1
|
Molins B, Rodríguez A, Llorenç V, Adán A. Biomaterial engineering strategies for modeling the Bruch's membrane in age-related macular degeneration. Neural Regen Res 2024; 19:2626-2636. [PMID: 38595281 PMCID: PMC11168499 DOI: 10.4103/nrr.nrr-d-23-01789] [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: 10/31/2023] [Revised: 01/03/2024] [Accepted: 02/06/2024] [Indexed: 04/11/2024] Open
Abstract
Age-related macular degeneration, a multifactorial inflammatory degenerative retinal disease, ranks as the leading cause of blindness in the elderly. Strikingly, there is a scarcity of curative therapies, especially for the atrophic advanced form of age-related macular degeneration, likely due to the lack of models able to fully recapitulate the native structure of the outer blood retinal barrier, the prime target tissue of age-related macular degeneration. Standard in vitro systems rely on 2D monocultures unable to adequately reproduce the structure and function of the outer blood retinal barrier, integrated by the dynamic interaction of the retinal pigment epithelium, the Bruch's membrane, and the underlying choriocapillaris. The Bruch's membrane provides structural and mechanical support and regulates the molecular trafficking in the outer blood retinal barrier, and therefore adequate Bruch's membrane-mimics are key for the development of physiologically relevant models of the outer blood retinal barrier. In the last years, advances in the field of biomaterial engineering have provided novel approaches to mimic the Bruch's membrane from a variety of materials. This review provides a discussion of the integrated properties and function of outer blood retinal barrier components in healthy and age-related macular degeneration status to understand the requirements to adequately fabricate Bruch's membrane biomimetic systems. Then, we discuss novel materials and techniques to fabricate Bruch's membrane-like scaffolds for age-related macular degeneration in vitro modeling, discussing their advantages and challenges with a special focus on the potential of Bruch's membrane-like mimics based on decellularized tissue.
Collapse
Affiliation(s)
- Blanca Molins
- Group of Ocular Inflammation: Clinical and Experimental Studies, Institut d’Investigacions Biomèdiques Agustí Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Andrea Rodríguez
- Group of Ocular Inflammation: Clinical and Experimental Studies, Institut d’Investigacions Biomèdiques Agustí Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Víctor Llorenç
- Group of Ocular Inflammation: Clinical and Experimental Studies, Institut d’Investigacions Biomèdiques Agustí Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Institut Clínic d’Oftalmologia (ICOF), Hospital Clínic Barcelona, Spain
| | - Alfredo Adán
- Group of Ocular Inflammation: Clinical and Experimental Studies, Institut d’Investigacions Biomèdiques Agustí Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Institut Clínic d’Oftalmologia (ICOF), Hospital Clínic Barcelona, Spain
| |
Collapse
|
2
|
Madrid M, Lakshmipathy U, Zhang X, Bharti K, Wall DM, Sato Y, Muschler G, Ting A, Smith N, Deguchi S, Kawamata S, Moore JC, Makovoz B, Sullivan S, Falco V, Al-Riyami AZ. Considerations for the development of iPSC-derived cell therapies: a review of key challenges by the JSRM-ISCT iPSC Committee. Cytotherapy 2024; 26:1382-1399. [PMID: 38958627 PMCID: PMC11471376 DOI: 10.1016/j.jcyt.2024.05.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 05/16/2024] [Accepted: 05/22/2024] [Indexed: 07/04/2024]
Abstract
Since their first production in 2007, human induced pluripotent stem cells (iPSCs) have provided a novel platform for the development of various cell therapies targeting a spectrum of diseases, ranging from rare genetic eye disorders to cancer treatment. However, several challenges must be tackled for iPSC-based cell therapy to enter the market and achieve broader global adoption. This white paper, authored by the Japanese Society for Regenerative Medicine (JSRM) - International Society for Cell Therapy (ISCT) iPSC Committee delves into the hurdles encountered in the pursuit of safe and economically viable iPSC-based therapies, particularly from the standpoint of the cell therapy industry. It discusses differences in global guidelines and regulatory frameworks, outlines a series of quality control tests required to ensure the safety of the cell therapy, and provides details and important considerations around cost of goods (COGs), including the impact of automated advanced manufacturing.
Collapse
Affiliation(s)
| | | | | | - Kapil Bharti
- National Eye Institute of the National Institutes of Health, Bethesda, USA
| | - Dominic M Wall
- Peter MacCallum Cancer Centre, Melbourne Australia; Cell Therapies Pty Ltd, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Yoji Sato
- National Institute of Health Sciences, Kawasaki, Japan
| | | | | | | | - Shuhei Deguchi
- CIRA Foundation, Facility for iPS Cell Therapy (FiT), Kyoto, Japan
| | - Shin Kawamata
- Cyto-Facto Inc., Kobe, Japan; Kobe University, Kobe, Japan.
| | | | | | | | | | - Arwa Z Al-Riyami
- Department of Hematology, Sultan Qaboos University Hospital, University Medical City, Muscat, Oman
| |
Collapse
|
3
|
Song HW, Solomon JN, Masri F, Mack A, Durand N, Cameau E, Dianat N, Hunter A, Oh S, Schoen B, Marsh M, Bravery C, Sumen C, Clarke D, Bharti K, Allickson JG, Lakshmipathy U. Bioprocessing considerations for generation of iPSCs intended for clinical application: perspectives from the ISCT Emerging Regenerative Medicine Technology working group. Cytotherapy 2024; 26:1275-1284. [PMID: 38970614 DOI: 10.1016/j.jcyt.2024.05.024] [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: 11/17/2023] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 07/08/2024]
Abstract
Approval of induced pluripotent stem cells (iPSCs) for the manufacture of cell therapies to support clinical trials is now becoming realized after 20 years of research and development. In 2022 the International Society for Cell and Gene Therapy (ISCT) established a Working Group on Emerging Regenerative Medicine Technologies, an area in which iPSCs-derived technologies are expected to play a key role. In this article, the Working Group surveys the steps that an end user should consider when generating iPSCs that are stable, well-characterised, pluripotent, and suitable for making differentiated cell types for allogeneic or autologous cell therapies. The objective is to provide the reader with a holistic view of how to achieve high-quality iPSCs from selection of the starting material through to cell banking. Key considerations include: (i) intellectual property licenses; (ii) selection of the raw materials and cell sources for creating iPSC intermediates and master cell banks; (iii) regulatory considerations for reprogramming methods; (iv) options for expansion in 2D vs. 3D cultures; and (v) available technologies and equipment for harvesting, washing, concentration, filling, cryopreservation, and storage. Some key process limitations are highlighted to help drive further improvement and innovation, and includes recommendations to close and automate current open and manual processes.
Collapse
Affiliation(s)
- Hannah W Song
- Center for Cellular Engineering, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | - Emmanuelle Cameau
- Cytiva, Pall Life Sciences 24-26 avenue de Winchester, CS5005, 78100 St. Germain-en-Laye, France
| | | | | | - Steve Oh
- Cellvec Pte. Ltd. 100 Pasir Panjang, #04-01/02, Singapore 118518 Singapore
| | - Brianna Schoen
- Charles River Laboratories Cell Solutions, Inc. 8500 Balboa Blvd. Suite 230 Northridge, CA 91320, USA
| | | | | | | | | | - Kapil Bharti
- National Eye Institute, National Institutes of Health, Bethsda, MD, USA
| | - Julie G Allickson
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
4
|
Liu H, Huang SS, Lingam G, Kai D, Su X, Liu Z. Advances in retinal pigment epithelial cell transplantation for retinal degenerative diseases. Stem Cell Res Ther 2024; 15:390. [PMID: 39482729 PMCID: PMC11526680 DOI: 10.1186/s13287-024-04007-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024] Open
Abstract
Retinal degenerative diseases are a leading cause of vision loss and blindness globally, impacting millions. These diseases result from progressive damage to retinal pigment epithelial (RPE) cells for which no curative or palliative treatments exist. Cell therapy, particularly RPE transplantation, has emerged as a promising strategy for vision restoration. This review provides a comprehensive overview of the recent advancements in clinical trials related to RPE transplantation. We discuss scaffold-free and scaffold-based approaches, including RPE cell suspensions and pre-organized RPE monolayers on biomaterial scaffolds. Key considerations, such as the form and preparation of RPE implants, delivery devices, strategies, and biodegradability of scaffolds, are examined. The article also explores the challenges and opportunities in RPE scaffold development, emphasising the crucial need for functional integration, immunomodulation, and long-term biocompatibility to ensure therapeutic efficacy. We also highlight ongoing efforts to optimise RPE transplantation methods and their potential to address retinal degenerative diseases.
Collapse
Affiliation(s)
- Hang Liu
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Centre for Innovation and Prevision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Suber S Huang
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Retina Center of Ohio, Cleveland, OH, USA
- Bascom Palmer Eye Institute, University of Miami, Coral Gables, FL, USA
| | - Gopal Lingam
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Centre for Innovation and Prevision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Ophthalmology, National University Hospital, Singapore, Singapore
- Singapore Eye Research Institute, Singapore, Singapore
| | - Dan Kai
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Xinyi Su
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Centre for Innovation and Prevision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Department of Ophthalmology, National University Hospital, Singapore, Singapore.
- Singapore Eye Research Institute, Singapore, Singapore.
| | - Zengping Liu
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Centre for Innovation and Prevision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Singapore Eye Research Institute, Singapore, Singapore.
| |
Collapse
|
5
|
Wu KY, Dhaliwal JK, Sasitharan A, Kalevar A. Cell Therapy for Retinal Degenerative Diseases: Progress and Prospects. Pharmaceutics 2024; 16:1299. [PMID: 39458628 PMCID: PMC11510658 DOI: 10.3390/pharmaceutics16101299] [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: 08/31/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Age-related macular degeneration (AMD) and retinitis pigmentosa (RP) are leading causes of vision loss, with AMD affecting older populations and RP being a rarer, genetically inherited condition. Both diseases result in progressive retinal degeneration, for which current treatments remain inadequate in advanced stages. This review aims to provide an overview of the retina's anatomy and physiology, elucidate the pathophysiology of AMD and RP, and evaluate emerging cell-based therapies for these conditions. Methods: A comprehensive review of the literature was conducted, focusing on cell therapy approaches, including embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), mesenchymal stem cells (MSCs), and retinal progenitor cells. Preclinical and clinical studies were analyzed to assess therapeutic potential, with attention to mechanisms such as cell replacement, neuroprotection, and paracrine effects. Relevant challenges, including ethical concerns and clinical translation, were also explored. Results: Cell-based therapies demonstrate potential for restoring retinal function and slowing disease progression through mechanisms like neuroprotection and cell replacement. Preclinical trials show promising outcomes, but clinical studies face significant hurdles, including challenges in cell delivery and long-term efficacy. Combination therapies integrating gene editing and biomaterials offer potential future advancements. Conclusions: While cell-based therapies for AMD and RP have made significant progress, substantial barriers to clinical application remain. Further research is essential to overcome these obstacles, improve delivery methods, and ensure the safe and effective translation of these therapies into clinical practice.
Collapse
Affiliation(s)
- Kevin Y. Wu
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada
| | - Jaskarn K. Dhaliwal
- Faculty of Health Sciences, Department of Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Akash Sasitharan
- Faculty of Medicine and Health Sciences, Department of Medicine, McGill University, Montreal, QC H3A 0GA, Canada
| | - Ananda Kalevar
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada
| |
Collapse
|
6
|
Rust R, Nih LR, Liberale L, Yin H, El Amki M, Ong LK, Zlokovic BV. Brain repair mechanisms after cell therapy for stroke. Brain 2024; 147:3286-3305. [PMID: 38916992 PMCID: PMC11449145 DOI: 10.1093/brain/awae204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/04/2024] [Accepted: 06/08/2024] [Indexed: 06/27/2024] Open
Abstract
Cell-based therapies hold great promise for brain repair after stroke. While accumulating evidence confirms the preclinical and clinical benefits of cell therapies, the underlying mechanisms by which they promote brain repair remain unclear. Here, we briefly review endogenous mechanisms of brain repair after ischaemic stroke and then focus on how different stem and progenitor cell sources can promote brain repair. Specifically, we examine how transplanted cell grafts contribute to improved functional recovery either through direct cell replacement or by stimulating endogenous repair pathways. Additionally, we discuss recently implemented preclinical refinement methods, such as preconditioning, microcarriers, genetic safety switches and universal (immune evasive) cell transplants, as well as the therapeutic potential of these pharmacologic and genetic manipulations to further enhance the efficacy and safety of cell therapies. By gaining a deeper understanding of post-ischaemic repair mechanisms, prospective clinical trials may be further refined to advance post-stroke cell therapy to the clinic.
Collapse
Affiliation(s)
- Ruslan Rust
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
| | - Lina R Nih
- Department of Brain Health, University of Nevada, Las Vegas, NV 89154, USA
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Hao Yin
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Mohamad El Amki
- Department of Neurology, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Lin Kooi Ong
- School of Health and Medical Sciences & Centre for Health Research, University of Southern Queensland, Toowoomba, QLD 4350, Australia
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
7
|
Novoa J, Westra I, Steeneveld E, Neves NF, Daleman L, Asensio AB, Davis RP, Carlotti F, Freund C, Rabelink T, Meij P, Wieles B. Validating human induced pluripotent stem cell-specific quality control tests for the release of an intermediate drug product in a Good Manufacturing Practice quality system. Cytotherapy 2024; 26:1105-1117. [PMID: 38703154 DOI: 10.1016/j.jcyt.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/29/2024] [Accepted: 04/10/2024] [Indexed: 05/06/2024]
Abstract
One of the challenges in Good Manufacturing Practice (GMP)-compliant human induced pluripotent stem cell (hiPSC) production is the validation of quality control (QC) tests specific for hiPSCs, which are required for GMP batch release. This study presents a comprehensive description of the validation process for hiPSC-specific GMP-compliant QC assays; more specifically, the validation of assays to assess the potential presence of residual episomal vectors (REVs), the expression of markers of the undifferentiated state and the directed differentiation potential of hiPSCs. Critical aspects and specific acceptance criteria were formulated in a validation plan prior to assay validation. Assay specificity, sensitivity and reproducibility were tested, and the equipment used for each assay was subjected to performance qualification. A minimum input of 20 000 cells (120 ng of genomic DNA) was defined for accurate determination of the presence of REVs. Furthermore, since vector loss in hiPSC lines is a passage-dependent process, we advocate screening for REVs between passages eight and 10, as testing at earlier passages might lead to unnecessary rejection of hiPSC lines. The cutoff value for assessment of markers of the undifferentiated state was set to the expression of at least three individual markers on at least 75% of the cells. When multi-color flow cytometry panels are used, a fluorescence minus one control is advised to ensure the control for fluorescent spread. For the assay to assess the directed differentiation potential, the detection limit was set to two of three positive lineage-specific markers for each of the three individual germ layers. All of our assays proved to be reproducible and specific. Our data demonstrate that our implemented analytical procedures are suitable as QC assays for the batch release of GMP-compliant hiPSCs.
Collapse
Affiliation(s)
- Juan Novoa
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Inge Westra
- Center for Cell and Gene Therapy, Leiden University Medical Center, Leiden, the Netherlands
| | - Esther Steeneveld
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Natascha Fonseca Neves
- Center for Cell and Gene Therapy, Leiden University Medical Center, Leiden, the Netherlands
| | - Lizanne Daleman
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Albert Blanch Asensio
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands; Novo Nordisk Foundation Center for Stem Cell Medicine, Leiden University Medical Center, the Netherlands
| | - Richard P Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands; Novo Nordisk Foundation Center for Stem Cell Medicine, Leiden University Medical Center, the Netherlands
| | - Françoise Carlotti
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Christian Freund
- Leiden University Medical Center hiPSC Hotel, Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ton Rabelink
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands; Novo Nordisk Foundation Center for Stem Cell Medicine, Leiden University Medical Center, the Netherlands.
| | - Pauline Meij
- Center for Cell and Gene Therapy, Leiden University Medical Center, Leiden, the Netherlands
| | - Brigitte Wieles
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
8
|
Hampton C, Bharti K, Song MJ. Tissue Engineering of Outer Blood Retina Barrier for Therapeutic Development. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2024; 31:100538. [PMID: 38962280 PMCID: PMC11218818 DOI: 10.1016/j.cobme.2024.100538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
Age related macular degeneration and other retinal degenerative disorders are characterized by disruption of the outer blood retinal barrier (oBRB) with subsequent ischemia, neovascularization, and atrophy. Despite the treatment advances, there remains no curative therapy, and no treatment targeted at regenerating native-like tissue for patients with late stages of the disease. Here we present advances in tissue engineering, focusing on bioprinting methods of generating tissue allowing for safe and reliable production of oBRB as well as tissue reprogramming with induced pluripotent stem cells for transplantation. We compare these approaches to organ-on-a-chip models for studying the dynamic nature of physiologic conditions. Highlighted within this review are studies that employ good manufacturing practices and use clinical grade methods that minimize potential risk to patients. Lastly, we illustrate recent clinical applications demonstrating both safety and efficacy for direct patient use. These advances provide an avenue for drug discovery and ultimately transplantation.
Collapse
Affiliation(s)
- Christopher Hampton
- Department of Ophthalmology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Kapil Bharti
- National Eye Institute, National Institute of Health, Bethesda, MD, USA
| | - Min Jae Song
- National Center for Advancing Translational Sciences, National Institute of Health, Rockville, MD, USA
| |
Collapse
|
9
|
Bellingrath JS, Li KV, Aziz K, Izzi JM, Liu YV, Singh MS. Large animal model species in pluripotent stem cell therapy research and development for retinal diseases: a systematic review. FRONTIERS IN OPHTHALMOLOGY 2024; 4:1377098. [PMID: 39253560 PMCID: PMC11381226 DOI: 10.3389/fopht.2024.1377098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 07/18/2024] [Indexed: 09/11/2024]
Abstract
Aim Retinal cell therapy modalities, in the category of advanced therapy medicinal products (ATMPs), are being developed to target several retinal diseases. Testing in large animal models (LAMs) is a crucial step in translating retinal ATMPs into clinical practice. However, challenges including budgetary and infrastructure constraints can hinder LAM research design and execution. Here, to facilitate the comparison of the various LAMs in pluripotent retinal cell therapy research, we aimed to systematically evaluate the species distribution, reported scientific utility, and methodology of a range of LAMs. Methods A systematic search using the words retina, stem cell, transplantation, large animal, pig, rabbit, dog, and nonhuman primate was conducted in the PubMed, Embase, Science Direct and GoogleScholar databases in February 2023. Results We included 22 studies involving pluripotent stem cells (induced pluripotent stem cells or human embryonic stem cells) in LAMs, including non-human primates (NHP), pigs, dogs, and rabbits. Nearly half of the studies utilized wild-type animal models. In other studies, retinal degeneration features were simulated via laser, chemical, or genetic insult. Transplants were delivered subretinally, either as cell suspensions or pre-formed monolayers (with or without biodegradable scaffolding). The transplanted cells dose per eye varied widely (40,000 - 4,000,000 per dose). Cells were delivered via vitrectomy surgery in 15 studies and by an "ab externo" approach in one study. Structural outcomes were assessed using confocal scanning laser ophthalmoscopy imaging. Functional outcomes included multifocal electroretinogram and, in one case, a measure of visual acuity. Generally, cell suspension transplants exhibited low intraretinal incorporation, while monolayer transplants incorporated more efficiently. Immune responses posed challenges for allogeneic transplants, suggesting that autologous iPSC-derived transplants may be required to decrease the likelihood of rejection. Conclusion The use of appropriate LAMs helps to advance the development of retinal ATMPs. The anatomical similarity of LAM and human eyes allows the implementation of clinically-relevant surgical techniques. While the FDA Modernization Act 2.0 has provided a framework to consider alternative methods including tissue-on-a-chip and human cell culture models for pharmacologic studies, LAM testing remains useful for cell and tissue replacement studies to inform the development of clinical trial protocols.
Collapse
Affiliation(s)
- Julia-Sophia Bellingrath
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Kang V Li
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kanza Aziz
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
| | - Jessica M Izzi
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ying V Liu
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mandeep S Singh
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
10
|
Deshmukh R, Sethi P, Singh B, Shiekmydeen J, Salave S, Patel RJ, Ali N, Rashid S, Elossaily GM, Kumar A. Recent Review on Biological Barriers and Host-Material Interfaces in Precision Drug Delivery: Advancement in Biomaterial Engineering for Better Treatment Therapies. Pharmaceutics 2024; 16:1076. [PMID: 39204421 PMCID: PMC11360117 DOI: 10.3390/pharmaceutics16081076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
Preclinical and clinical studies have demonstrated that precision therapy has a broad variety of treatment applications, making it an interesting research topic with exciting potential in numerous sectors. However, major obstacles, such as inefficient and unsafe delivery systems and severe side effects, have impeded the widespread use of precision medicine. The purpose of drug delivery systems (DDSs) is to regulate the time and place of drug release and action. They aid in enhancing the equilibrium between medicinal efficacy on target and hazardous side effects off target. One promising approach is biomaterial-assisted biotherapy, which takes advantage of biomaterials' special capabilities, such as high biocompatibility and bioactive characteristics. When administered via different routes, drug molecules deal with biological barriers; DDSs help them overcome these hurdles. With their adaptable features and ample packing capacity, biomaterial-based delivery systems allow for the targeted, localised, and prolonged release of medications. Additionally, they are being investigated more and more for the purpose of controlling the interface between the host tissue and implanted biomedical materials. This review discusses innovative nanoparticle designs for precision and non-personalised applications to improve precision therapies. We prioritised nanoparticle design trends that address heterogeneous delivery barriers, because we believe intelligent nanoparticle design can improve patient outcomes by enabling precision designs and improving general delivery efficacy. We additionally reviewed the most recent literature on biomaterials used in biotherapy and vaccine development, covering drug delivery, stem cell therapy, gene therapy, and other similar fields; we have also addressed the difficulties and future potential of biomaterial-assisted biotherapies.
Collapse
Affiliation(s)
- Rohitas Deshmukh
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, India;
| | - Pranshul Sethi
- Department of Pharmacology, College of Pharmacy, Shri Venkateshwara University, Gajraula 244236, India;
| | - Bhupendra Singh
- School of Pharmacy, Graphic Era Hill University, Dehradun 248002, India;
- Department of Pharmacy, S.N. Medical College, Agra 282002, India
| | | | - Sagar Salave
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, India;
| | - Ravish J. Patel
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, Changa, Anand 388421, India;
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia;
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia;
| | - Gehan M. Elossaily
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia;
| | - Arun Kumar
- School of Pharmacy, Sharda University, Greater Noida 201310, India
| |
Collapse
|
11
|
Liu H, Lu S, Chen M, Gao N, Yang Y, Hu H, Ren Q, Liu X, Chen H, Zhu Q, Li S, Su J. Towards Stem/Progenitor Cell-Based Therapies for Retinal Degeneration. Stem Cell Rev Rep 2024; 20:1459-1479. [PMID: 38809490 DOI: 10.1007/s12015-024-10740-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Abstract
Retinal degeneration (RD) is a leading cause of blindness worldwide and includes conditions such as retinitis pigmentosa (RP), age-related macular degeneration (AMD), and Stargardt's disease (STGD). These diseases result in the permanent loss of vision due to the progressive and irreversible degeneration of retinal cells, including photoreceptors (PR) and the retinal pigment epithelium (RPE). The adult human retina has limited abilities to regenerate and repair itself, making it challenging to achieve complete self-replenishment and functional repair of retinal cells. Currently, there is no effective clinical treatment for RD. Stem cell therapy, which involves transplanting exogenous stem cells such as retinal progenitor cells (RPCs), embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and mesenchymal stem cells (MSCs), or activating endogenous stem cells like Müller Glia (MG) cells, holds great promise for regenerating and repairing retinal cells in the treatment of RD. Several preclinical and clinical studies have shown the potential of stem cell-based therapies for RD. However, the clinical translation of these therapies for the reconstruction of substantial vision still faces significant challenges. This review provides a comprehensive overview of stem/progenitor cell-based therapy strategies for RD, summarizes recent advances in preclinical studies and clinical trials, and highlights the major challenges in using stem/progenitor cell-based therapies for RD.
Collapse
Affiliation(s)
- Hui Liu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Shuaiyan Lu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Ming Chen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Na Gao
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yuhe Yang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Huijuan Hu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Qing Ren
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiaoyu Liu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Hongxu Chen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Qunyan Zhu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325011, China
| | - Shasha Li
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, 325001, China.
| | - Jianzhong Su
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325011, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, 325001, China.
| |
Collapse
|
12
|
Nair PP, Keskar MP, Borghare PT, Dzoagbe HY, Kumar T. The New Era of Therapeutic Strategies for the Management of Retinitis Pigmentosa: A Narrative Review of the Pathomolecular Mechanism for Gene Therapies. Cureus 2024; 16:e66814. [PMID: 39280562 PMCID: PMC11393205 DOI: 10.7759/cureus.66814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 08/11/2024] [Indexed: 09/18/2024] Open
Abstract
Retinitis pigmentosa, or RP, is a group of inherited retinal degenerations involving progressive loss of photoreceptor cells- rods and cones- ultimately causing severe vision loss and blindness. RP, although a very common ailment, continues to be an incurable disease with little to be done medically. However, with the breakthroughs in gene therapy and stem cell transplantation in recent years, a new door has been opened to the treatment of RP. This narrative review summarizes the pathomolecular mechanisms of RP, focusing on the genetic and molecular abnormalities that lead to the process of retinal degeneration. In this section, we talk about the current theories of how RP develops, gene mutations, oxidative stress, and inflammation. We also delve into new therapeutic approaches such as gene therapy, stem cell transplantation and genome surgery, which are designed to either replace or repair the damaged photoreceptors to restore vision and ultimately enhance the life of the RP patient. Another topic covered is the obstacles and research frontiers of these revolutionary treatments. This article is intended to give a complete overview of the molecular processes of RP and the promising treatment strategies that could change the way this devastating disease is treated.
Collapse
Affiliation(s)
- Praveena P Nair
- Otolaryngology, Mandsaur Institute of Ayurved Education and Research, Bhunyakhedi, IND
- Otolaryngology, Parul Institute of Ayurved, Parul University, Limda, IND
| | - Manjiri P Keskar
- Otolaryngology, Parul institute of Ayurved, Parul University, Limda, IND
| | - Pramod T Borghare
- Otolaryngology, Mahatma Gandhi Ayurved College Hospital and Research, Wardha, IND
| | - Hellen Y Dzoagbe
- Anatomy, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Tanish Kumar
- Medicine, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
13
|
Chucair-Elliott AJ, Ocañas SR, Pham K, Machalinski A, Plafker S, Stout MB, Elliott MH, Freeman WM. Age- and sex- divergent translatomic responses of the mouse retinal pigmented epithelium. Neurobiol Aging 2024; 140:41-59. [PMID: 38723422 PMCID: PMC11173338 DOI: 10.1016/j.neurobiolaging.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/18/2024]
Abstract
Aging is the main risk factor for age-related macular degeneration (AMD), a retinal neurodegenerative disease that leads to irreversible blindness, particularly in people over 60 years old. Retinal pigmented epithelium (RPE) atrophy is an AMD hallmark. Genome-wide chromatin accessibility, DNA methylation, and gene expression studies of AMD and control RPE demonstrate epigenomic/transcriptomic changes occur during AMD onset and progression. However, mechanisms by which molecular alterations of normal aging impair RPE function and contribute to AMD pathogenesis are unclear. Here, we specifically interrogate the RPE translatome with advanced age and across sexes in a novel RPE reporter mouse model. We find differential age- and sex- associated transcript expression with overrepresentation of pathways related to inflammation in the RPE. Concordant with impaired RPE function, the phenotypic changes in the aged translatome suggest that aged RPE becomes immunologically active, in both males and females, with some sex-specific signatures, which supports the need for sex representation for in vivo studies.
Collapse
Affiliation(s)
- Ana J Chucair-Elliott
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
| | - Sarah R Ocañas
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Kevin Pham
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Adeline Machalinski
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Scott Plafker
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Michael B Stout
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Michael H Elliott
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Willard M Freeman
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
14
|
Ghosh S, Sharma R, Bammidi S, Koontz V, Nemani M, Yazdankhah M, Kedziora KM, Stolz DB, Wallace CT, Yu-Wei C, Franks J, Bose D, Shang P, Ambrosino HM, Dutton JR, Geng Z, Montford J, Ryu J, Rajasundaram D, Hose S, Sahel JA, Puertollano R, Finkel T, Zigler JS, Sergeev Y, Watkins SC, Goetzman ES, Ferrington DA, Flores-Bellver M, Kaarniranta K, Sodhi A, Bharti K, Handa JT, Sinha D. The AKT2/SIRT5/TFEB pathway as a potential therapeutic target in non-neovascular AMD. Nat Commun 2024; 15:6150. [PMID: 39034314 PMCID: PMC11271488 DOI: 10.1038/s41467-024-50500-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 07/10/2024] [Indexed: 07/23/2024] Open
Abstract
Non-neovascular or dry age-related macular degeneration (AMD) is a multi-factorial disease with degeneration of the aging retinal-pigmented epithelium (RPE). Lysosomes play a crucial role in RPE health via phagocytosis and autophagy, which are regulated by transcription factor EB/E3 (TFEB/E3). Here, we find that increased AKT2 inhibits PGC-1α to downregulate SIRT5, which we identify as an AKT2 binding partner. Crosstalk between SIRT5 and AKT2 facilitates TFEB-dependent lysosomal function in the RPE. AKT2/SIRT5/TFEB pathway inhibition in the RPE induced lysosome/autophagy signaling abnormalities, disrupted mitochondrial function and induced release of debris contributing to drusen. Accordingly, AKT2 overexpression in the RPE caused a dry AMD-like phenotype in aging Akt2 KI mice, as evident from decline in retinal function. Importantly, we show that induced pluripotent stem cell-derived RPE encoding the major risk variant associated with AMD (complement factor H; CFH Y402H) express increased AKT2, impairing TFEB/TFE3-dependent lysosomal function. Collectively, these findings suggest that targeting the AKT2/SIRT5/TFEB pathway may be an effective therapy to delay the progression of dry AMD.
Collapse
Affiliation(s)
- Sayan Ghosh
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ruchi Sharma
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sridhar Bammidi
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Victoria Koontz
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mihir Nemani
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Meysam Yazdankhah
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Katarzyna M Kedziora
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Donna Beer Stolz
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Callen T Wallace
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Cheng Yu-Wei
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jonathan Franks
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Devika Bose
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | - James R Dutton
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Zhaohui Geng
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Jair Montford
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jiwon Ryu
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stacey Hose
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - José-Alain Sahel
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Institut De La Vision, INSERM, CNRS, Sorbonne Université, Paris, France
| | - Rosa Puertollano
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Toren Finkel
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - J Samuel Zigler
- Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yuri Sergeev
- Protein Biochemistry & Molecular Modeling Group, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Simon C Watkins
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eric S Goetzman
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Deborah A Ferrington
- Doheny Eye Institute, Pasadena, CA, USA
- Department of Ophthalmology, University of California Los Angeles, Los Angeles, CA, USA
| | - Miguel Flores-Bellver
- Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kai Kaarniranta
- Department of Ophthalmology, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
- Department of Molecular Genetics, University of Lodz, Lodz, Poland
| | - Akrit Sodhi
- Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kapil Bharti
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institutes of Health, Bethesda, MD, USA.
| | - James T Handa
- Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Debasish Sinha
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
15
|
Herardot E, Liboz M, Lamour G, Malo M, Plancheron A, Habeler W, Geiger C, Frank E, Campillo C, Monville C, Ben M'Barek K. Biomechanical Characterization of Retinal Pigment Epitheliums Derived from hPSCs Using Atomic Force Microscopy. Stem Cell Rev Rep 2024; 20:1340-1352. [PMID: 38627341 PMCID: PMC11222240 DOI: 10.1007/s12015-024-10717-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2024] [Indexed: 07/04/2024]
Abstract
The retinal pigment epithelium (RPE), a multifunctional cell monolayer located at the back of the eye, plays a crucial role in the survival and homeostasis of photoreceptors. Dysfunction or death of RPE cells leads to retinal degeneration and subsequent vision loss, such as in Age-related macular degeneration and some forms of Retinitis Pigmentosa. Therefore, regenerative medicine that aims to replace RPE cells by new cells obtained from the differentiation of human pluripotent stem cells, is the focus of intensive research. However, despite their critical interest in therapy, there is a lack of biomechanical RPE surface description. Such biomechanical properties are tightly related to their functions. Herein, we used atomic force microscopy (AFM) to analyze both the structural and mechanical properties of RPEs obtained from four cell lines and at different stages of epithelial formation. To characterize epitheliums, we used apical markers in immunofluorescence and showed the increase of transepithelial resistance, as well as the ability to secrete cytokines with an apico-basal polarity. Then, we used AFM to scan the apical surface of living or fixed RPE cells. We show that RPE monolayers underwent softening of apical cell center as well as stiffening of cell borders over epithelial formation. We also observed apical protrusions that depend on actin network, suggesting the formation of microvilli at the surface of RPE epitheliums. These RPE cell characteristics are essential for their functions into the retina and AFM studies may improve the characterization of the RPE epithelium suitable for cell therapy.
Collapse
Affiliation(s)
- Elise Herardot
- Université Paris-Saclay, Univ Evry, INSERM, IStem, UMR861, 91100, Corbeil-Essonnes, France
| | - Maxime Liboz
- Université Paris-Saclay, Univ Evry, CY Cergy Paris Université, CNRS, LAMBE, 91025, Evry-Courcouronnes, France
| | - Guillaume Lamour
- Université Paris-Saclay, Univ Evry, CY Cergy Paris Université, CNRS, LAMBE, 91025, Evry-Courcouronnes, France
| | - Michel Malo
- Université Paris-Saclay, Univ Evry, CY Cergy Paris Université, CNRS, LAMBE, 91025, Evry-Courcouronnes, France
| | - Alexandra Plancheron
- Université Paris-Saclay, Univ Evry, INSERM, IStem, UMR861, 91100, Corbeil-Essonnes, France
- IStem, CECS, 91100, Corbeil-Essonnes, France
| | - Walter Habeler
- Université Paris-Saclay, Univ Evry, INSERM, IStem, UMR861, 91100, Corbeil-Essonnes, France
- IStem, CECS, 91100, Corbeil-Essonnes, France
| | - Camille Geiger
- Université Paris-Saclay, Univ Evry, INSERM, IStem, UMR861, 91100, Corbeil-Essonnes, France
- IStem, CECS, 91100, Corbeil-Essonnes, France
| | - Elie Frank
- Université Paris-Saclay, Univ Evry, INSERM, IStem, UMR861, 91100, Corbeil-Essonnes, France
| | - Clément Campillo
- Université Paris-Saclay, Univ Evry, CY Cergy Paris Université, CNRS, LAMBE, 91025, Evry-Courcouronnes, France
- Institut Universitaire de France (IUF), Paris, France
| | - Christelle Monville
- Université Paris-Saclay, Univ Evry, INSERM, IStem, UMR861, 91100, Corbeil-Essonnes, France.
| | - Karim Ben M'Barek
- Université Paris-Saclay, Univ Evry, INSERM, IStem, UMR861, 91100, Corbeil-Essonnes, France.
- IStem, CECS, 91100, Corbeil-Essonnes, France.
| |
Collapse
|
16
|
Perez VL, Mousa HM, Miyagishima KJ, Reed AA, Su AJA, Greenwell TN, Washington KM. Retinal transplant immunology and advancements. Stem Cell Reports 2024; 19:817-829. [PMID: 38729155 PMCID: PMC11297553 DOI: 10.1016/j.stemcr.2024.04.007] [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: 06/07/2023] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
Several gaps and barriers remain for transplanting stem cells into the eye to treat ocular disease, especially diseases of the retina. While the eye has historically been considered immune privileged, recent thinking has identified the immune system as both a barrier and an opportunity for eye stem cell transplantation. Recent approaches leveraging scaffolds or cloaking have been considered in other tissues beyond immune suppression. This perspective paper outlines approaches for transplantation and proposes opportunities to overcome barriers of the immune system in stem cell transplantation in the eye.
Collapse
Affiliation(s)
- Victor L Perez
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA; Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Hazem M Mousa
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA
| | | | - Amberlynn A Reed
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - An-Jey A Su
- Department of Surgery, University of Colorado School of Medicine, CU Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Thomas N Greenwell
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kia M Washington
- Department of Surgery, University of Colorado School of Medicine, CU Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
17
|
Novoa JJ, Westra IM, Steeneveld E, Fonseca Neves N, Arendzen CH, Rajaei B, Grundeken E, Yildiz M, van der Valk W, Salvador A, Carlotti F, Dijkers PF, Locher H, van den Berg CW, Raymond KI, Kirkeby A, Mummery CL, Rabelink TJ, Freund C, Meij P, Wieles B. Good Manufacturing Practice-compliant human induced pluripotent stem cells: from bench to putative clinical products. Cytotherapy 2024; 26:556-566. [PMID: 38483359 DOI: 10.1016/j.jcyt.2024.02.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND AIMS Few human induced pluripotent stem cell (hiPSC) lines are Good Manufacturing Practice (GMP)-compliant, limiting the clinical use of hiPSC-derived products. Here, we addressed this by establishing and validating an in-house platform to produce GMP-compliant hiPSCs that would be appropriate for producing both allogeneic and autologous hiPSC-derived products. METHODS Our standard research protocol for hiPSCs production was adapted and translated into a GMP-compliant platform. In addition to the generation of GMP-compliant hiPSC, the platform entails the methodology for donor recruitment, consent and screening, donor material procurement, hiPSCs manufacture, in-process control, specific QC test validation, QC testing, product release, hiPSCs storage and stability testing. For platform validation, one test run and three production runs were performed. Highest-quality lines were selected to establish master cell banks (MCBs). RESULTS Two MCBs were successfully released under GMP conditions. They demonstrated safety (sterility, negative mycoplasma, endotoxins <5.0 EU/mL and negative adventitious agents), cell identity (>75% of cells expressing markers of undifferentiated state, identical STR profile, normal karyotype in >20 metaphases), purity (negative residual vectors and no plasmid integration in the genome) and potency (expression of at least two of the three markers for each of the three germ layers). In addition, directed differentiation to somitoids (skeletal muscle precursors) and six potential clinical products from all three germ layers was achieved: pancreatic islets (endoderm), kidney organoids and cardiomyocytes (mesoderm), and keratinocytes, GABAergic interneurons and inner-ear organoids (ectoderm). CONCLUSIONS We successfully developed and validated a platform for generating GMP-compliant hiPSC lines. The two MCBs released were shown to differentiate into clinical products relevant for our own and other regenerative medicine interests.
Collapse
Affiliation(s)
- Juan J Novoa
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Inge M Westra
- Center for Cell and Gene Therapy, Leiden University Medical Center, Leiden, The Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Esther Steeneveld
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Natascha Fonseca Neves
- Center for Cell and Gene Therapy, Leiden University Medical Center, Leiden, The Netherlands
| | - Christiaan H Arendzen
- LUMC hiPSC Hotel, Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bahareh Rajaei
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Esmée Grundeken
- LUMC hiPSC Hotel, Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Mehmet Yildiz
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Wouter van der Valk
- OtoBiology Leiden, Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, The Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Alison Salvador
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, Copenhagen, Denmark
| | - Françoise Carlotti
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Pascale F Dijkers
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Heiko Locher
- OtoBiology Leiden, Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, The Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Cathelijne W van den Berg
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Karine I Raymond
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Agnete Kirkeby
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, Copenhagen, Denmark
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Ton J Rabelink
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands.
| | - Christian Freund
- LUMC hiPSC Hotel, Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Pauline Meij
- Center for Cell and Gene Therapy, Leiden University Medical Center, Leiden, The Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Brigitte Wieles
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
18
|
Jiang B, Wang X, Rivera-Bolanos N, Ameer GA. Generation of Autologous Vascular Endothelial Cells for Patients with Peripheral Artery Disease. J Cardiovasc Transl Res 2024; 17:558-569. [PMID: 37861912 DOI: 10.1007/s12265-023-10452-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/10/2023] [Indexed: 10/21/2023]
Abstract
Peripheral artery disease (PAD) is a prevalent cardiovascular disease with risks of limb loss. Our objective is to establish an autologous cell source for vascular regeneration to achieve limb salvage in PAD. Six PAD patients (age 50-80) were enrolled with their peripheral blood collected to derive vascular endothelial cells (ECs) with two different approaches: (1) endothelial progenitor cell (EPC) approach and (2) induced pluripotent stem cell (iPSC) approach. The iPSC approach successfully generated patient-specific ECs for all PAD patients, while the EPC approach did not yield any colony-forming ECs in any of the patients. The patient-derived iPSC-ECs expressed endothelial markers and exhibited endothelial functions. However, elevated inflammatory status with VCAM-1 expression was observed in the patient-derived cells. Pharmacological treatment with resveratrol resulted in patient-specific responses in cell viability and VCAM-1 expression. Our study demonstrates the potential of iPSC-ECs for autologous regenerative therapy in PAD, offering promise for personalized treatments for ischemic PAD.
Collapse
Affiliation(s)
- Bin Jiang
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA.
- Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Rd, Tech B382, Evanston, IL, 60208, USA.
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| | - Xinlong Wang
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
- Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Rd, Tech B382, Evanston, IL, 60208, USA
| | - Nancy Rivera-Bolanos
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
- Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Rd, Tech B382, Evanston, IL, 60208, USA
| | - Guillermo A Ameer
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA.
- Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Rd, Tech B382, Evanston, IL, 60208, USA.
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA.
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, 60208, USA.
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
19
|
Trousil J, Cabral JV, Voukali E, Nováčková J, Pop-Georgievski O, Vacík T, Studený P, Studenovska H, Jirsova K. Electrospun poly(l-lactide- co-dl-lactide) nanofibrous scaffold as substrate for ex vivo limbal epithelial cell cultivation. Heliyon 2024; 10:e30970. [PMID: 38803982 PMCID: PMC11128869 DOI: 10.1016/j.heliyon.2024.e30970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/13/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024] Open
Abstract
Ultrathin electrospun poly (l-lactide-co-dl-lactide) nanofibrous membranes coated with fibronectin were explored as scaffolds for the ex vivo cultivation of limbal epithelial cells (LECs) for the treatment of limbal stem cell deficiency. The developed scaffolds were compared with the "gold-standard" fibrin gel. The resulting membranes composed of nanofibers possessed a very low thickness of 4 μm and allowed very good optical transparency in the wet state. The fibronectin-coated nanofibrous scaffolds demonstrated LEC expansion and successful cultivation similar to that on fibrin gel. Unlike the regular cobblestone epithelial cell morphology on the fibrin gel, the nanofibrous scaffold presented a mostly irregular epithelial morphology with a shift to a mesenchymal phenotype, as confirmed by the upregulation of profibroblastic genes: ACTA2 (p = 0.023), FBLN1 (p < 0.001), and THY1 (p < 0.001). Both culture conditions revealed comparable expression of stem cell markers, including KLF4, ΔNp63α and ABCG2, emphasizing the promise of polylactide-based nanofibrous membranes for further investigations.
Collapse
Affiliation(s)
- Jiří Trousil
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, Czech Republic
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Joao Victor Cabral
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Eleni Voukali
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Jitka Nováčková
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Ognen Pop-Georgievski
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Tomáš Vacík
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Pavel Studený
- Ophthalmology Department, Third Faculty of Medicine, Charles University and University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Hana Studenovska
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Katerina Jirsova
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| |
Collapse
|
20
|
Zhang Y, Shi Y, Khan MM, Xiao F, Chen W, Tao W, Yao K, Kong N. Ocular RNA nanomedicine: engineered delivery nanoplatforms in treating eye diseases. Trends Biotechnol 2024:S0167-7799(24)00121-5. [PMID: 38821834 DOI: 10.1016/j.tibtech.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/23/2024] [Accepted: 05/06/2024] [Indexed: 06/02/2024]
Abstract
Ocular disorders remain a major global health challenge with unmet medical needs. RNA nanomedicine has shown significant therapeutic benefits and safety profiles in patients with complex eye disorders, already benefiting numerous patients with gene-related eye disorders. The effective delivery of RNA to the unique structure of the eye is challenging owing to RNA instability, off-target effects, and ocular physiological barriers. Specifically tailored RNA medication, coupled with sophisticated engineered delivery platforms, is crucial to guide and advance developments in treatments for oculopathy. Herein we review recent advances in RNA-based nanomedicine, innovative delivery strategies, and current clinical progress and present challenges in ocular disease therapy.
Collapse
Affiliation(s)
- Yiming Zhang
- Liangzhu Laboratory, Zhejiang University Medical Center and Zhejiang Provincial Key Lab of Ophthalmology, Eye Center of The Second Affiliated Hospital, Zhejiang University, Hangzhou, China; Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yesi Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Muhammad M Khan
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Fan Xiao
- Liangzhu Laboratory, Zhejiang University Medical Center and Zhejiang Provincial Key Lab of Ophthalmology, Eye Center of The Second Affiliated Hospital, Zhejiang University, Hangzhou, China; Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wei Chen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Ke Yao
- Liangzhu Laboratory, Zhejiang University Medical Center and Zhejiang Provincial Key Lab of Ophthalmology, Eye Center of The Second Affiliated Hospital, Zhejiang University, Hangzhou, China.
| | - Na Kong
- Liangzhu Laboratory, Zhejiang University Medical Center and Zhejiang Provincial Key Lab of Ophthalmology, Eye Center of The Second Affiliated Hospital, Zhejiang University, Hangzhou, China; Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
21
|
Shechter Y, Cohen R, Namestnikov M, Shapira A, Barak A, Barzelay A, Dvir T. Sequential Fabrication of a Three-Layer Retina-like Structure. Gels 2024; 10:336. [PMID: 38786253 PMCID: PMC11121616 DOI: 10.3390/gels10050336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 04/24/2024] [Accepted: 05/12/2024] [Indexed: 05/25/2024] Open
Abstract
Tissue engineering is considered a promising approach to treating advanced degenerative maculopathies such as nonexudative age-related macular degeneration (AMD), the leading cause of blindness worldwide. The retina consists of several hierarchical tissue layers, each of which is supported by a layer underneath. Each of these layers has a different morphology and requires distinct conditions for proper assembly. In fact, a prerequisite step for the assembly of each of these layers is the organization of the layer underneath. Advanced retinal degeneration includes degeneration of the other retina layers, including the choroid, the retinal pigmented epithelium (RPE), and the photoreceptors. Here, we report a step-by-step fabrication process of a three-layer retina-like structure. The process included the 3D printing of a choroid-like structure in an extracellular matrix (ECM) hydrogel, followed by deposition of the RPE monolayer. After the formation of the blood vessel-RPE interface, the photoreceptor cells were deposited to interact with the RPE layer. At the end of the fabrication process, each layer was characterized for its morphology and expression of specific markers, and the integration of the three-layer retina was evaluated. We envision that such a retina-like structure may be able to attenuate the deterioration of a degenerated retina and improve engraftment and regeneration. This retinal implant may potentially be suitable for a spectrum of macular degenerative diseases for which there are currently no cures and may save millions from complete blindness.
Collapse
Affiliation(s)
- Yahel Shechter
- Shmunis School of Biomedicine and Cancer Research, Faculty of Life Science, Tel Aviv University, Tel Aviv 6997801, Israel; (Y.S.); (R.C.); (M.N.); (A.S.)
| | - Roni Cohen
- Shmunis School of Biomedicine and Cancer Research, Faculty of Life Science, Tel Aviv University, Tel Aviv 6997801, Israel; (Y.S.); (R.C.); (M.N.); (A.S.)
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Michael Namestnikov
- Shmunis School of Biomedicine and Cancer Research, Faculty of Life Science, Tel Aviv University, Tel Aviv 6997801, Israel; (Y.S.); (R.C.); (M.N.); (A.S.)
| | - Assaf Shapira
- Shmunis School of Biomedicine and Cancer Research, Faculty of Life Science, Tel Aviv University, Tel Aviv 6997801, Israel; (Y.S.); (R.C.); (M.N.); (A.S.)
| | - Adiel Barak
- Division of Ophthalmology, Tel Aviv Medical Center, Tel Aviv 6423906, Israel;
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Aya Barzelay
- Division of Ophthalmology, Tel Aviv Medical Center, Tel Aviv 6423906, Israel;
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tal Dvir
- Shmunis School of Biomedicine and Cancer Research, Faculty of Life Science, Tel Aviv University, Tel Aviv 6997801, Israel; (Y.S.); (R.C.); (M.N.); (A.S.)
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol Center for Regenerative Biotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
22
|
Kajita K, Nishida M, Kurimoto Y, Yokota S, Sugita S, Semba T, Shirae S, Hayashi N, Ozaki A, Miura Y, Maeda A, Mitamura Y, Takahashi M, Mandai M. Graft cell expansion from hiPSC-RPE strip after transplantation in primate eyes with or without RPE damage. Sci Rep 2024; 14:10044. [PMID: 38698112 PMCID: PMC11065889 DOI: 10.1038/s41598-024-60895-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 04/29/2024] [Indexed: 05/05/2024] Open
Abstract
Clinical studies using suspensions or sheets of human pluripotent cell-derived retinal pigment epithelial cells (hiPSC-RPE) have been conducted globally for diseases such as age-related macular degeneration. Despite being minimally invasive, cell suspension transplantation faces challenges in targeted cell delivery and frequent cell leakage. Conversely, although the RPE sheet ensures targeted delivery with correct cell polarity, it requires invasive surgery, and graft preparation is time-consuming. We previously reported hiPSC-RPE strips as a form of quick cell aggregate that allows for reliable cell delivery to the target area with minimal invasiveness. In this study, we used a microsecond pulse laser to create a local RPE ablation model in cynomolgus monkey eyes. The hiPSC-RPE strips were transplanted into the RPE-ablated and intact sites. The hiPSC-RPE strip stably survived in all transplanted monkey eyes. The expansion area of the RPE from the engrafted strip was larger at the RPE injury site than at the intact site with no tumorigenic growth. Histological observation showed a monolayer expansion of the transplanted RPE cells with the expression of MERTK apically and collagen type 4 basally. The hiPSC-RPE strip is considered a beneficial transplantation option for RPE cell therapy.
Collapse
Affiliation(s)
- Keisuke Kajita
- Kobe City Eye Hospital, 2-1-8, Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
- Department of Ophthalmology, Institute of Biomedical Sciences, Graduate School, Tokushima University, Tokushima, Japan
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, 2-2-3, Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
- Vision Care Inc. Kobe Eye Center 5F, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Mitsuhiro Nishida
- Kobe City Eye Hospital, 2-1-8, Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, 2-2-3, Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Yasuo Kurimoto
- Kobe City Eye Hospital, 2-1-8, Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, 2-2-3, Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Satoshi Yokota
- Kobe City Eye Hospital, 2-1-8, Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, 2-2-3, Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Sunao Sugita
- Kobe City Eye Hospital, 2-1-8, Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, 2-2-3, Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
- Vision Care Inc. Kobe Eye Center 5F, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Toshika Semba
- Vision Care Inc. Kobe Eye Center 5F, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Satoshi Shirae
- Vision Care Inc. Kobe Eye Center 5F, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Naoko Hayashi
- Vision Care Inc. Kobe Eye Center 5F, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Atsuta Ozaki
- Kobe City Eye Hospital, 2-1-8, Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
- Department of Ophthalmology, Mie University Graduate School of Medicine, Tsu, 514-8507, Japan
| | - Yoko Miura
- Institute of Biomedical Optics, University of Lübeck, Peter-Monnik-Weg 4, 23562, Lübeck, Germany
- Department of Ophthalmology, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Akiko Maeda
- Kobe City Eye Hospital, 2-1-8, Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, 2-2-3, Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Yoshinori Mitamura
- Department of Ophthalmology, Institute of Biomedical Sciences, Graduate School, Tokushima University, Tokushima, Japan
| | - Masayo Takahashi
- Kobe City Eye Hospital, 2-1-8, Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
- Vision Care Inc. Kobe Eye Center 5F, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Michiko Mandai
- Kobe City Eye Hospital, 2-1-8, Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, 2-2-3, Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.
| |
Collapse
|
23
|
Cerneckis J, Cai H, Shi Y. Induced pluripotent stem cells (iPSCs): molecular mechanisms of induction and applications. Signal Transduct Target Ther 2024; 9:112. [PMID: 38670977 PMCID: PMC11053163 DOI: 10.1038/s41392-024-01809-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 03/09/2024] [Accepted: 03/17/2024] [Indexed: 04/28/2024] Open
Abstract
The induced pluripotent stem cell (iPSC) technology has transformed in vitro research and holds great promise to advance regenerative medicine. iPSCs have the capacity for an almost unlimited expansion, are amenable to genetic engineering, and can be differentiated into most somatic cell types. iPSCs have been widely applied to model human development and diseases, perform drug screening, and develop cell therapies. In this review, we outline key developments in the iPSC field and highlight the immense versatility of the iPSC technology for in vitro modeling and therapeutic applications. We begin by discussing the pivotal discoveries that revealed the potential of a somatic cell nucleus for reprogramming and led to successful generation of iPSCs. We consider the molecular mechanisms and dynamics of somatic cell reprogramming as well as the numerous methods available to induce pluripotency. Subsequently, we discuss various iPSC-based cellular models, from mono-cultures of a single cell type to complex three-dimensional organoids, and how these models can be applied to elucidate the mechanisms of human development and diseases. We use examples of neurological disorders, coronavirus disease 2019 (COVID-19), and cancer to highlight the diversity of disease-specific phenotypes that can be modeled using iPSC-derived cells. We also consider how iPSC-derived cellular models can be used in high-throughput drug screening and drug toxicity studies. Finally, we discuss the process of developing autologous and allogeneic iPSC-based cell therapies and their potential to alleviate human diseases.
Collapse
Affiliation(s)
- Jonas Cerneckis
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Hongxia Cai
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Yanhong Shi
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA.
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA.
| |
Collapse
|
24
|
Faura G, Studenovska H, Sekac D, Ellederova Z, Petrovski G, Eide L. The Effects of the Coating and Aging of Biodegradable Polylactic Acid Membranes on In Vitro Primary Human Retinal Pigment Epithelium Cells. Biomedicines 2024; 12:966. [PMID: 38790928 PMCID: PMC11117638 DOI: 10.3390/biomedicines12050966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
Age-related macular degeneration (AMD) is the most frequent cause of blindness in developed countries. The replacement of dysfunctional human retinal pigment epithelium (hRPE) cells by the transplantation of in vitro-cultivated hRPE cells to the affected area emerges as a feasible strategy for regenerative therapy. Synthetic biomimetic membranes arise as powerful hRPE cell carriers, but as biodegradability is a requirement, it also poses a challenge due to its limited durability. hRPE cells exhibit several characteristics that putatively respond to the type of membrane carrier, and they can be used as biomarkers to evaluate and further optimize such membranes. Here, we analyze the pigmentation, transepithelial resistance, genome integrity, and maturation markers of hRPE cells plated on commercial polycarbonate (PC) versus in-house electrospun polylactide-based (PLA) membranes, both enabling separate apical/basolateral compartments. Our results show that PLA is superior to PC-based membranes for the cultivation of hRPEs, and the BEST1/RPE65 maturation markers emerge as the best biomarkers for addressing the quality of hRPE cultivated in vitro. The stability of the cultures was observed to be affected by PLA aging, which is an effect that could be partially palliated by the coating of the PLA membranes.
Collapse
Affiliation(s)
- Georgina Faura
- Department of Medical Biochemistry, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- CIDETEC, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San Sebastián, Spain
| | - Hana Studenovska
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, 162 00 Prague, Czech Republic;
| | - David Sekac
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, 277 21 Libechov, Czech Republic; (D.S.); (Z.E.)
- Department of Cell Biology, Faculty of Science, Charles University, 128 00 Prague, Czech Republic
| | - Zdenka Ellederova
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, 277 21 Libechov, Czech Republic; (D.S.); (Z.E.)
| | - Goran Petrovski
- Center for Eye Research and Innovative Diagnostics, Department of Ophthalmology, Oslo University Hospital and Institute for Clinical Medicine, University of Oslo, 0424 Oslo, Norway;
- Norwegian Center for Stem Cell Research, Oslo University Hospital, 0424 Oslo, Norway
- Department of Ophthalmology, University Hospital Centre, University of Split School of Medicine, 21000 Split, Croatia
- UKLO Network, University St. Kliment Ohridski, 7000 Bitola, North Macedonia
| | - Lars Eide
- Department of Medical Biochemistry, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Department of Medical Biochemistry, Oslo University Hospital, 0424 Oslo, Norway
| |
Collapse
|
25
|
Xu M, Gao Y, Yin W, Liu Q, Yuan S. RNA-sequencing expression profile and functional analysis of retinal pigment epithelium in atrophic age-related macular degeneration. J Biomed Res 2024; 38:1-12. [PMID: 38808557 PMCID: PMC11461538 DOI: 10.7555/jbr.37.20230320] [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: 01/03/2024] [Revised: 03/11/2024] [Accepted: 03/19/2024] [Indexed: 05/30/2024] Open
Abstract
The retinal pigment epithelium (RPE) is fundamental to sustaining retinal homeostasis. RPE abnormality leads to visual defects and blindness, including age-related macular degeneration (AMD). Although breakthroughs have been made in the treatment of neovascular AMD, effective intervention for atrophic AMD is largely absent. The inadequate knowledge of RPE pathology is hindered by a lack of patient RPE datasets, especially at the single-cell resolution. In this study, we delved into a large-scale single-cell resource of AMD donors in which RPE cells were occupied in a substantial proportion. Bulk RNA-seq datasets of atrophic AMD were integrated to extract molecular characteristics of RPE in the pathogenesis of atrophic AMD. Both in vivo and in vitro models revealed that carboxypeptidase X, M14 family member 2 (CPXM2) was specifically expressed in the RPE cells of atrophic AMD, which might be induced by oxidative stress and involved in the epithelial-mesenchymal transition of RPE cells. Additionally, silencing of CPXM2 inhibited the mesenchymal phenotype of RPE cells in an oxidative stress cell model. Thus, our results demonstrate that CPXM2 plays a crucial role in regulating atrophic AMD and may serve as a potential therapeutic target for atrophic AMD.
Collapse
Affiliation(s)
- Miao Xu
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yan Gao
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Wenjie Yin
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Qinghuai Liu
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Songtao Yuan
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
26
|
McCall MA. Pig Models in Retinal Research and Retinal Disease. Cold Spring Harb Perspect Med 2024; 14:a041296. [PMID: 37553210 PMCID: PMC10982707 DOI: 10.1101/cshperspect.a041296] [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] [Indexed: 08/10/2023]
Abstract
The pig has been used as a large animal model in biomedical research for many years and its use continues to increase because induced mutations phenocopy several inherited human diseases. In addition, they are continuous breeders, can be propagated by artificial insemination, have large litter sizes (on the order of mice), and can be genetically manipulated using all of the techniques that are currently available in mice. The pioneering work of Petters and colleagues set the stage for the use of the pig as a model of inherited retinal disease. In the last 10 years, the pig has become a model of choice where specific disease-causing mutations that are not phenocopied in rodents need to be studied and therapeutic approaches explored. The pig is not only used for retinal eye disease but also for the study of the cornea and lens. This review attempts to show how broad the use of the pig has become and how it has contributed to the assessment of treatments for eye disease. In the last 10 years, there have been several reviews that included the use of the pig in biomedical research (see body of the review) that included information about retinal disease. None directly discuss the use of the pig as an animal model for retinal diseases, including inherited diseases, where a single genetic mutation has been identified or for multifactorial diseases such as glaucoma and diabetic retinopathy. Although the pig is used to explore diseases of the cornea and lens, this review focuses on how and why the pig, as a large animal model, is useful for research in neural retinal disease and its treatment.
Collapse
Affiliation(s)
- Maureen A McCall
- Departments of Ophthalmology & Visual Sciences and Anatomical Sciences & Neurobiology, University of Louisville, Louisville, Kentucky 40202, USA
| |
Collapse
|
27
|
Baqué-Vidal L, Main H, Petrus-Reurer S, Lederer AR, Beri NE, Bär F, Metzger H, Zhao C, Efstathopoulos P, Saietz S, Wrona A, Jaberi E, Willenbrock H, Reilly H, Hedenskog M, Moussaud-Lamodière E, Kvanta A, Villaescusa JC, La Manno G, Lanner F. Clinically compliant cryopreservation of differentiated retinal pigment epithelial cells. Cytotherapy 2024; 26:340-350. [PMID: 38349309 DOI: 10.1016/j.jcyt.2024.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 04/07/2024]
Abstract
BACKGROUND AIMS Age-related macular degeneration (AMD) is the most common cause of blindness in elderly patients within developed countries, affecting more than 190 million worldwide. In AMD, the retinal pigment epithelial (RPE) cell layer progressively degenerates, resulting in subsequent loss of photoreceptors and ultimately vision. There is currently no cure for AMD, but therapeutic strategies targeting the complement system are being developed to slow the progression of the disease. METHODS Replacement therapy with pluripotent stem cell-derived (hPSC) RPEs is an alternative treatment strategy. A cell therapy product must be produced in accordance with Good Manufacturing Practices at a sufficient scale to facilitate extensive pre-clinical and clinical testing. Cryopreservation of the final cell product is therefore highly beneficial, as the manufacturing, pre-clinical and clinical testing can be separated in time and location. RESULTS We found that mature hPSC-RPE cells do not survive conventional cryopreservation techniques. However, replating the cells 2-5 days before cryopreservation facilitates freezing. The replated and cryopreserved hPSC-RPE cells maintained their identity, purity and functionality as characteristic RPEs, shown by cobblestone morphology, pigmentation, transcriptional profile, RPE markers, transepithelial resistance and pigment epithelium-derived factor secretion. Finally, we showed that the optimal replating time window can be tracked noninvasively by following the change in cobblestone morphology. CONCLUSIONS The possibility of cryopreserving the hPSC-RPE product has been instrumental in our efforts in manufacturing and performing pre-clinical testing with the aim for clinical translation.
Collapse
Affiliation(s)
- Laura Baqué-Vidal
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Heather Main
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Sandra Petrus-Reurer
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden; Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden; Department of Surgery, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Alex R Lederer
- Laboratory of Neurodevelopmental Systems Biology, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Nefeli-Eirini Beri
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Frederik Bär
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Hugo Metzger
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Cheng Zhao
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | | | - Sarah Saietz
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | | | - Elham Jaberi
- Cell Therapy R&D, Novo Nordisk A/S, Måløv, Denmark
| | | | - Hazel Reilly
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Mona Hedenskog
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Elisabeth Moussaud-Lamodière
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Anders Kvanta
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | | | - Gioele La Manno
- Laboratory of Neurodevelopmental Systems Biology, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Fredrik Lanner
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden; Ming Wai Lau Center for Reparative Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
28
|
Iezzi R, Scruggs BA, Gandhi J, Zenti FN, Shafi N, Berger A, Marmorstein AD. Early Choriocapillaris Loss in a Porcine Model of RPE Cell Debridement Precedes Pathology That Simulates Advanced Macular Degeneration. Invest Ophthalmol Vis Sci 2024; 65:8. [PMID: 38568620 PMCID: PMC10996981 DOI: 10.1167/iovs.65.4.8] [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: 10/19/2023] [Accepted: 03/11/2024] [Indexed: 04/05/2024] Open
Abstract
Purpose No large-mammal surgical models exist for geographic atrophy (GA), choroidal neovascularization (CNV), and pachychoroidal vascular remodeling. Our goal was to develop a porcine RPE debridement model of advanced macular degeneration to study photoreceptor cell loss and choroidal remodeling. Methods Seven 2-month-old female domestic pigs were used for this study. After 25G vitrectomy, the area centralis was detached via subretinal bleb. A nitinol wire (Finesse Flex Loop) was used to debride RPE cells across a 3- to 5-mm diameter region. Fluid-air exchange was performed, and 20% SF6 gas injected. Animals underwent fundus photography, fluorescein angiography, optical coherence tomography (OCT), and OCT-angiography (OCTA) at 2 weeks, 1 month, 2 months, 3 months, and 6 months postoperatively. Retinal histology was obtained at euthanasia, 2 months (n = 3), 3 months (n = 2), or 6 months (n = 2) after surgery. Results RPE debridement resulted in GA with rapid loss of choriocapillaris, progressive loss of photoreceptors, and pachychoroidal changes in Sattler's and Haller's layers in all seven eyes undergoing debridement within 2 months. OCT and histological findings included subretinal disciform scar with overlying outer retinal atrophy; outer retinal tubulations and subretinal hyper-reflective material. OCTA revealed type 2 CNV (n = 4) at the edges of the debridement zone by 2 months, but there was no significant exudation noted at any time point. Conclusions Surgical debridement of the RPE results in GA, CNV, and pachychoroid and reproduced all forms of advanced macular degeneration. This surgical model may be useful in examining the role of RPE and other cell replacement in treating advanced macular disease.
Collapse
Affiliation(s)
- Raymond Iezzi
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, United States
| | - Brittni A. Scruggs
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, United States
| | - Jarel Gandhi
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, United States
| | - Francesca N. Zenti
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, United States
| | - Noah Shafi
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, United States
| | - Aubrey Berger
- Regenerative Sciences, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, United States
| | - Alan D. Marmorstein
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
29
|
Hosseini SM, Borys B, Karimi-Abdolrezaee S. Neural stem cell therapies for spinal cord injury repair: an update on recent preclinical and clinical advances. Brain 2024; 147:766-793. [PMID: 37975820 DOI: 10.1093/brain/awad392] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/22/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023] Open
Abstract
Traumatic spinal cord injury (SCI) is a leading cause of lifelong disabilities. Permanent sensory, motor and autonomic impairments after SCI are substantially attributed to degeneration of spinal cord neurons and axons, and disintegration of neural network. To date, minimal regenerative treatments are available for SCI with an unmet need for new therapies to reconstruct the damaged spinal cord neuron-glia network and restore connectivity with the supraspinal pathways. Multipotent neural precursor cells (NPCs) have a unique capacity to generate neurons, oligodendrocytes and astrocytes. Due to this capacity, NPCs have been an attractive cell source for cellular therapies for SCI. Transplantation of NPCs has been extensively tested in preclinical models of SCI in the past two decades. These studies have identified opportunities and challenges associated with NPC therapies. While NPCs have the potential to promote neuroregeneration through various mechanisms, their low long-term survival and integration within the host injured spinal cord limit the functional benefits of NPC-based therapies for SCI. To address this challenge, combinatorial strategies have been developed to optimize the outcomes of NPC therapies by enriching SCI microenvironment through biomaterials, genetic and pharmacological therapies. In this review, we will provide an in-depth discussion on recent advances in preclinical NPC-based therapies for SCI. We will discuss modes of actions and mechanism by which engrafted NPCs contribute to the repair process and functional recovery. We will also provide an update on current clinical trials and new technologies that have facilitated preparation of medical-grade human NPCs suitable for transplantation in clinical studies.
Collapse
Affiliation(s)
- Seyed Mojtaba Hosseini
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba Winnipeg, Manitoba R3E 0J9, Canada
- Manitoba Multiple Sclerosis Research Center, Winnipeg, Manitoba R3E 0J9, Canada
| | - Ben Borys
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba Winnipeg, Manitoba R3E 0J9, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba Winnipeg, Manitoba R3E 0J9, Canada
- Manitoba Multiple Sclerosis Research Center, Winnipeg, Manitoba R3E 0J9, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba R3E 3P4, Canada
| |
Collapse
|
30
|
Bose D, Ortolan D, Farnoodian M, Sharma R, Bharti K. Considerations for Developing an Autologous Induced Pluripotent Stem Cell (iPSC)-Derived Retinal Pigment Epithelium (RPE) Replacement Therapy. Cold Spring Harb Perspect Med 2024; 14:a041295. [PMID: 37487631 PMCID: PMC10910357 DOI: 10.1101/cshperspect.a041295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Cell-replacement therapies are a new class of treatments, which include induced pluripotent stem cell (iPSC)-derived tissues that aim to replace degenerated cells. iPSCs can potentially be used to generate any cell type of the body, making them a powerful tool for treating degenerative diseases. Cell replacement for retinal degenerative diseases is at the forefront of cell therapies, given the accessibility of the eye for surgical procedures and a huge unmet medical need for retinal degenerative diseases with no current treatment options. Clinical trials are ongoing in different parts of the world using stem cell-derived retinal pigment epithelium (RPE). This review focuses on scientific and regulatory considerations when developing an iPSC-derived RPE cell therapy from the development of a robust and efficient differentiation protocol to critical quality control assays for cell validation, the choice of an appropriate animal model for preclinical testing, and the regulatory aspects that dictate the final approval for proceeding to a first-in-human clinical trial.
Collapse
Affiliation(s)
- Devika Bose
- Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Davide Ortolan
- Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Mitra Farnoodian
- Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Ruchi Sharma
- Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Kapil Bharti
- Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
31
|
Barone F, Bunea I, Creel K, Sharma R, Amaral J, Maminishkis A, Bharti K. An Automated Visual Psychophysics Method to Measure Visual Function in Swine Preclinical Animal Model. Transl Vis Sci Technol 2024; 13:8. [PMID: 38470318 PMCID: PMC10941991 DOI: 10.1167/tvst.13.3.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 01/28/2024] [Indexed: 03/13/2024] Open
Abstract
Purpose The aim of this study was to develop and validate a test to assess visual function in pigs using the visual psychophysics contrast sensitivity function. Methods We utilized a touchscreen along with a pellet reward dispenser to train three Göttingen pigs on a visual psychophysics test and determined their contrast sensitivity function. Images with different contrast resolutions were used as visual stimuli and presented against a control image in a two-choice test. Following animals' acclimatization and the first phase of training, the system was arranged such that animals could self-run multiple consecutive trials without human intervention. Results All animals were trained within a week and remembered the task with 1 day of reinforcement when tested 1 month after the last visual assessment. All trained animals performed well during the trial with minimal screen side bias, especially at contrast threshold above 40%. Conclusions Göttingen pigs are trainable for a visual psychophysics test and able to self-run the trial without human intervention. Translational Relevance Contrast sensitivity is one of the key parameters to assess visual function in humans. The possibility of measuring the same parameters in a large animal model allows for a better translation and understanding of drug safety and efficacy in preclinical ophthalmology.
Collapse
Affiliation(s)
- Francesca Barone
- Ocular and Stem Cell Translational Research Section, National Eye Institute, NIH, Bethesda, MD, USA
| | - Irina Bunea
- Ocular and Stem Cell Translational Research Section, National Eye Institute, NIH, Bethesda, MD, USA
| | - Kristi Creel
- Ocular and Stem Cell Translational Research Section, National Eye Institute, NIH, Bethesda, MD, USA
| | - Ruchi Sharma
- Ocular and Stem Cell Translational Research Section, National Eye Institute, NIH, Bethesda, MD, USA
| | - Juan Amaral
- Ocular and Stem Cell Translational Research Section, National Eye Institute, NIH, Bethesda, MD, USA
| | - Arvydas Maminishkis
- Translational Research Core, Ophthalmic Genetics and Visual Function Branch, National Eye Institute, NIH, Bethesda, MD, USA
| | - Kapil Bharti
- Ocular and Stem Cell Translational Research Section, National Eye Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
32
|
Wang B, Starr AL, Fraser HB. Cell-type-specific cis-regulatory divergence in gene expression and chromatin accessibility revealed by human-chimpanzee hybrid cells. eLife 2024; 12:RP89594. [PMID: 38358392 PMCID: PMC10942608 DOI: 10.7554/elife.89594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024] Open
Abstract
Although gene expression divergence has long been postulated to be the primary driver of human evolution, identifying the genes and genetic variants underlying uniquely human traits has proven to be quite challenging. Theory suggests that cell-type-specific cis-regulatory variants may fuel evolutionary adaptation due to the specificity of their effects. These variants can precisely tune the expression of a single gene in a single cell-type, avoiding the potentially deleterious consequences of trans-acting changes and non-cell type-specific changes that can impact many genes and cell types, respectively. It has recently become possible to quantify human-specific cis-acting regulatory divergence by measuring allele-specific expression in human-chimpanzee hybrid cells-the product of fusing induced pluripotent stem (iPS) cells of each species in vitro. However, these cis-regulatory changes have only been explored in a limited number of cell types. Here, we quantify human-chimpanzee cis-regulatory divergence in gene expression and chromatin accessibility across six cell types, enabling the identification of highly cell-type-specific cis-regulatory changes. We find that cell-type-specific genes and regulatory elements evolve faster than those shared across cell types, suggesting an important role for genes with cell-type-specific expression in human evolution. Furthermore, we identify several instances of lineage-specific natural selection that may have played key roles in specific cell types, such as coordinated changes in the cis-regulation of dozens of genes involved in neuronal firing in motor neurons. Finally, using novel metrics and a machine learning model, we identify genetic variants that likely alter chromatin accessibility and transcription factor binding, leading to neuron-specific changes in the expression of the neurodevelopmentally important genes FABP7 and GAD1. Overall, our results demonstrate that integrative analysis of cis-regulatory divergence in chromatin accessibility and gene expression across cell types is a promising approach to identify the specific genes and genetic variants that make us human.
Collapse
Affiliation(s)
- Ban Wang
- Department of Biology, Stanford UniversityStanfordUnited States
| | | | - Hunter B Fraser
- Department of Biology, Stanford UniversityStanfordUnited States
| |
Collapse
|
33
|
Liu YV, Li KV, Li Z, Lu Y, McNally MM, Esposito EP, Aziz K, Singh MS. Transpupillary-Guided Trans-Scleral Transplantation of Subretinal Grafts in a Retinal Degeneration Mouse Model. J Vis Exp 2024:10.3791/65448. [PMID: 38345250 PMCID: PMC11294988 DOI: 10.3791/65448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2024] Open
Abstract
Transplantation of photoreceptor cells and retinal pigment epithelial (RPE) cells provide a potential therapy for retinal degeneration diseases. Subretinal transplantation of therapeutic donor cells into mouse recipients is challenging due to the limited surgical space allowed by the small volume of the mouse eye. We developed a trans-scleral surgical transplantation platform with direct transpupillary vision guidance to facilitate the subretinal delivery of exogenous cells in mouse recipients. The platform was tested using retinal cell suspensions and three-dimensional retinal sheets collected from rod-rich Rho::EGFP mice and cone-rich OPN1LW-EGFP;NRL-/- mice, respectively. Live/dead assay showed low cell mortality for both forms of donor cells. Retinal grafts were successfully delivered into the subretinal space of a mouse model of retinal degeneration, Rd1/NS, with minimum surgical complications as detected by multimodal confocal scanning laser ophthalmoscope (cSLO) imaging. Two months post-transplantation, histological staining demonstrated evidence of advanced maturation of the retinal grafts into 'adult' rods and cones (by robust Rho::EGFP, S-opsin, and OPN1LW:EGFP expression, respectively) in the subretinal space. Here, we provide a surgical platform that can enable highly accurate subretinal delivery with a low rate of complications in mouse recipients. This technique offers precision and relative ease of skill acquisition. Furthermore, the technique could be used not only for studies of subretinal cell transplantation but also for other intraocular therapeutic studies including gene therapies.
Collapse
Affiliation(s)
- Ying V Liu
- Wilmer Eye Institute, Johns Hopkins University School of Medicine
| | - Kang V Li
- Wilmer Eye Institute, Johns Hopkins University School of Medicine
| | - Zhuolin Li
- Wilmer Eye Institute, Johns Hopkins University School of Medicine
| | - Yuchen Lu
- Wilmer Eye Institute, Johns Hopkins University School of Medicine
| | - Minda M McNally
- Wilmer Eye Institute, Johns Hopkins University School of Medicine
| | | | - Kanza Aziz
- Wilmer Eye Institute, Johns Hopkins University School of Medicine
| | - Mandeep S Singh
- Wilmer Eye Institute, Johns Hopkins University School of Medicine; Department of Genetic Medicine, Johns Hopkins University School of Medicine;
| |
Collapse
|
34
|
Wang W, Yang T, Chen S, Liang L, Wang Y, Ding Y, Xiong W, Ye X, Guo Y, Shen S, Chen H, Chen J. Tissue engineering RPE sheet derived from hiPSC-RPE cell spheroids supplemented with Y-27632 and RepSox. J Biol Eng 2024; 18:7. [PMID: 38229139 DOI: 10.1186/s13036-024-00405-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 01/08/2024] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Retinal pigment epithelium (RPE) cell therapy is a promising way to treat many retinal diseases. However, obtaining transplantable RPE cells is time-consuming and less effective. This study aimed to develop novel strategies for generating engineered RPE patches with physiological characteristics. RESULTS Our findings revealed that RPE cells derived from human induced pluripotent stem cells (hiPSCs) successfully self-assembled into spheroids. The RPE spheroids treated with Y27632 and Repsox had increased expression of epithelial markers and RPE-specific genes, along with improved cell viability and barrier function. Transcriptome analysis indicated enhanced cell adhesion and extracellular matrix (ECM) organization in RPE spheroids. These RPE spheroids could be seeded and bioprinted on collagen vitrigel (CV) membranes to construct engineered RPE sheets. Circular RPE patches, obtained by trephining a specific section of the RPE sheet, exhibited abundant microvilli and pigment particles, as well as reduced proliferative capacity and enhanced maturation. CONCLUSIONS Our study suggests that the supplementation of small molecules and 3D spheroid culture, as well as the bioprinting technique, can be effective methods to promote RPE cultivation and construct engineered RPE sheets, which may support future clinical RPE cell therapy and the development of RPE models for research applications.
Collapse
Grants
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
- NSFC-RGC, 32061160469, N_CUHK432/20 National Natural Science Foundation of China
Collapse
Affiliation(s)
- Wenxuan Wang
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Tingting Yang
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Sihui Chen
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Liying Liang
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Yingxin Wang
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Yin Ding
- The University of Hong Kong - Shenzhen Hospital, Shenzhen, China
| | - Wei Xiong
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| | - Xiuhong Ye
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Yonglong Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Shuhao Shen
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Hang Chen
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Jiansu Chen
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China.
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China.
- Aier Eye Institute, Changsha, Hunan, China.
| |
Collapse
|
35
|
Esposito EP, Han IC, Johnson TV. Gene and cell-based therapies for retinal and optic nerve disease. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:243-262. [PMID: 39341657 DOI: 10.1016/b978-0-323-90120-8.00016-2] [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: 10/01/2024]
Abstract
Leading causes of blindness worldwide include neurodegenerative diseases of the retina, which cause irreversible loss of retinal pigment epithelium (RPE) and photoreceptors, and optic neuropathies, which result in retinal ganglion cell (RGC) death. Because photoreceptor and RGCs do not spontaneously regenerate in mammals, including humans, vision loss from these conditions is, at present, permanent. Recent advances in gene and cell-based therapies have provided new hope to patients affected by these conditions. This chapter reviews the current state and future of these approaches to treating ocular neurodegenerative disease. Gene therapies for retinal degeneration and optic neuropathies primarily focus on correcting known pathogenic mutations that cause inherited conditions to halt progression. There are multiple retinal and optic neuropathy gene therapies in clinical trials, and one retinal gene therapy is approved in the United States, Canada, Europe, and Australia. Cell-based therapies are mutation agnostic and have the potential to repopulate neurons regardless of the underlying etiology of degeneration. While photoreceptor cell replacement is nearing a human clinical trial, RPE transplantation is currently in phase I/II clinical trials. RGC replacement faces numerous logistical challenges, but preclinical research has laid the foundation for functional repair of optic neuropathies to be feasible.
Collapse
Affiliation(s)
- Edward P Esposito
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ian C Han
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Thomas V Johnson
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
36
|
Dujardin C, Habeler W, Monville C, Letourneur D, Simon-Yarza T. Advances in the engineering of the outer blood-retina barrier: From in-vitro modelling to cellular therapy. Bioact Mater 2024; 31:151-177. [PMID: 37637086 PMCID: PMC10448242 DOI: 10.1016/j.bioactmat.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/13/2023] [Accepted: 08/06/2023] [Indexed: 08/29/2023] Open
Abstract
The outer blood-retina barrier (oBRB), crucial for the survival and the proper functioning of the overlying retinal layers, is disrupted in numerous diseases affecting the retina, leading to the loss of the photoreceptors and ultimately of vision. To study the oBRB and/or its degeneration, many in vitro oBRB models have been developed, notably to investigate potential therapeutic strategies against retinal diseases. Indeed, to this day, most of these pathologies are untreatable, especially once the first signs of degeneration are observed. To cure those patients, a current strategy is to cultivate in vitro a mature oBRB epithelium on a custom membrane that is further implanted to replace the damaged native tissue. After a description of the oBRB and the related diseases, this review presents an overview of the oBRB models, from the simplest to the most complex. Then, we propose a discussion over the used cell types, for their relevance to study or treat the oBRB. Models designed for in vitro applications are then examined, by paying particular attention to the design evolution in the last years, the development of pathological models and the benefits of co-culture models, including both the retinal pigment epithelium and the choroid. Lastly, this review focuses on the models developed for in vivo implantation, with special emphasis on the choice of the material, its processing and its characterization, before discussing the reported pre-clinical and clinical trials.
Collapse
Affiliation(s)
- Chloé Dujardin
- Université Paris Cité, Université Sorbonne Paris Nord, Laboratory for Vascular Translational Science (LVTS) INSERM-U1148, 75018 Paris, France
| | - Walter Habeler
- INSERM U861, I-Stem, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 91100, Corbeil-Essonnes, France
- U861, I-Stem, AFM, Université Paris-Saclay, Université D’Evry, 91100, Corbeil-Essonnes, France
- CECS, Centre D’étude des Cellules Souches, 91100, Corbeil-Essonnes, France
| | - Christelle Monville
- INSERM U861, I-Stem, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 91100, Corbeil-Essonnes, France
- U861, I-Stem, AFM, Université Paris-Saclay, Université D’Evry, 91100, Corbeil-Essonnes, France
| | - Didier Letourneur
- Université Paris Cité, Université Sorbonne Paris Nord, Laboratory for Vascular Translational Science (LVTS) INSERM-U1148, 75018 Paris, France
| | - Teresa Simon-Yarza
- Université Paris Cité, Université Sorbonne Paris Nord, Laboratory for Vascular Translational Science (LVTS) INSERM-U1148, 75018 Paris, France
| |
Collapse
|
37
|
Liu Q, Liu J, Guo M, Sung TC, Wang T, Yu T, Tian Z, Fan G, Wu W, Higuchi A. Comparison of retinal degeneration treatment with four types of different mesenchymal stem cells, human induced pluripotent stem cells and RPE cells in a rat retinal degeneration model. J Transl Med 2023; 21:910. [PMID: 38098048 PMCID: PMC10720187 DOI: 10.1186/s12967-023-04785-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/05/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Retinal degeneration (RD) is a group of disorders on irreversible vision loss. Multiple types of stem cells were used in clinical trials for RD treatment. However, it remains unknown what kinds of stem cells are most effective for the treatment. Therefore, we investigated the subretinal transplantation of several types of stem cells, human adipose-derived stem cells (hADSCs), amniotic fluid stem cells (hAFSCs), bone marrow stem cells (hBMSCs), dental pulp stem cells (hDPSCs), induced pluripotent stem cell (hiPSC), and hiPSC-derived retinal pigment epithelium (RPE) cells for protection effects, paracrine effects and treatment efficiency in an RD disease model rats. METHODS The generation and characterization of these stem cells and hiPSC-derived RPE cells were performed before transplantation. The stem cells or hiPSC-derived RPE cell suspension labelled with CellTracker Green to detect transplanted cells were delivered into the subretinal space of 3-week-old RCS rats. The control group received subretinal PBS injection or non-injection. A series of detections including fundus photography, optomotor response (OMR) evaluations, light-dark box testing, electroretinography (ERG), and hematoxylin and eosin (HE) staining of retinal sections were conducted after subretinal injection of the cells. RESULTS Each stem cell, hiPSC-derived RPE cell or PBS (blank experiment) was successfully transplanted into at least six RCS rats subretinally. Compared with the control rats, RCS rats subjected to subretinal transplantation of any stem cells except hiPSCs showed higher ERG waves (p < 0.05) and quantitative OMR (qOMR) index values (hADSCs: 1.166, hAFSCs: 1.249, hBMSCs: 1.098, hDPSCs: 1.238, hiPSCs: 1.208, hiPSC-RPE cells: 1.294, non-injection: 1.03, PBS: 1.06), which indicated better visual function, at 4 weeks post-injection. However, only rats that received hiPSC-derived RPE cells maintained their visual function at 8 weeks post-injection (p < 0.05). The outer nuclear layer thickness observed in histological sections after HE staining showed the same pattern as the ERG and qOMR results. CONCLUSIONS Compared to hiPSC-derived RPE cells, adult and fetal stem cells yielded improvements in visual function for up to 4 weeks post-injection; this outcome was mainly based on the paracrine effects of several types of growth factors secreted by the stem cells. Patients with RD will benefit from the stem cell therapy.
Collapse
Affiliation(s)
- Qian Liu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, 325027, Zhejiang, China
| | - Jun Liu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, 325027, Zhejiang, China
| | - Minmei Guo
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, 325027, Zhejiang, China
| | - Tzu-Cheng Sung
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, 325027, Zhejiang, China
| | - Ting Wang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, 325027, Zhejiang, China
| | - Tao Yu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, 325027, Zhejiang, China
| | - Zeyu Tian
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, 325027, Zhejiang, China
| | - Guoping Fan
- Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210, Shanghai, China
| | - Wencan Wu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, 325027, Zhejiang, China
| | - Akon Higuchi
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, 325027, Zhejiang, China.
- Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan, 32001, Taiwan.
| |
Collapse
|
38
|
Farjood F, Manos JD, Wang Y, Williams AL, Zhao C, Borden S, Alam N, Prusky G, Temple S, Stern JH, Boles NC. Identifying biomarkers of heterogeneity and transplantation efficacy in retinal pigment epithelial cells. J Exp Med 2023; 220:e20230913. [PMID: 37728563 PMCID: PMC10510736 DOI: 10.1084/jem.20230913] [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: 05/26/2023] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 09/21/2023] Open
Abstract
Transplantation of retinal pigment epithelial (RPE) cells holds great promise for patients with retinal degenerative diseases, such as age-related macular degeneration. In-depth characterization of RPE cell product identity and critical quality attributes are needed to enhance efficacy and safety of replacement therapy strategies. Here, we characterized an adult RPE stem cell-derived (RPESC-RPE) cell product using bulk and single-cell RNA sequencing (scRNA-seq), assessing functional cell integration in vitro into a mature RPE monolayer and in vivo efficacy by vision rescue in the Royal College of Surgeons rats. scRNA-seq revealed several distinct subpopulations in the RPESC-RPE product, some with progenitor markers. We identified RPE clusters expressing genes associated with in vivo efficacy and increased cell integration capability. Gene expression analysis revealed lncRNA (TREX) as a predictive marker of in vivo efficacy. TREX knockdown decreased cell integration while overexpression increased integration in vitro and improved vision rescue in the RCS rats.
Collapse
Affiliation(s)
| | | | - Yue Wang
- Neural Stem Cell Institute, Rensselaer, NY, USA
| | | | | | | | - Nazia Alam
- Burke Neurological Institute at Weill Cornell Medicine, White Plains, NY, USA
| | - Glen Prusky
- Burke Neurological Institute at Weill Cornell Medicine, White Plains, NY, USA
| | | | | | | |
Collapse
|
39
|
Lee IK, Xie R, Luz-Madrigal A, Min S, Zhu J, Jin J, Edwards KL, Phillips MJ, Ludwig AL, Gamm DM, Gong S, Ma Z. Micromolded honeycomb scaffold design to support the generation of a bilayered RPE and photoreceptor cell construct. Bioact Mater 2023; 30:142-153. [PMID: 37575875 PMCID: PMC10415596 DOI: 10.1016/j.bioactmat.2023.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/27/2023] [Accepted: 07/22/2023] [Indexed: 08/15/2023] Open
Abstract
Age-related macular degeneration (AMD) causes blindness due to loss of retinal pigment epithelium (RPE) and photoreceptors (PRs), which comprise the two outermost layers of the retina. Given the small size of the macula and the importance of direct contact between RPE and PRs, the use of scaffolds for targeted reconstruction of the outer retina in later stage AMD and other macular dystrophies is particularly attractive. We developed microfabricated, honeycomb-patterned, biodegradable poly(glycerol sebacate) (PGS) scaffolds to deliver organized, adjacent layers of RPE and PRs to the subretinal space. Furthermore, an optimized process was developed to photocure PGS, shortening scaffold production time from days to minutes. The resulting scaffolds robustly supported the seeding of human pluripotent stem cell-derived RPE and PRs, either separately or as a dual cell-layered construct. These advanced, economical, and versatile scaffolds can accelerate retinal cell transplantation efforts and benefit patients with AMD and other retinal degenerative diseases.
Collapse
Affiliation(s)
- In-Kyu Lee
- Department of Electrical and Computer Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Ruosen Xie
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Agustin Luz-Madrigal
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Comparative Biomedical Sciences, University of Wisconsin–Madison, Madison, WI, 53706, USA
| | - Seunghwan Min
- Department of Electrical and Computer Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Jingcheng Zhu
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Jiahe Jin
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | | | - M. Joseph Phillips
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Allison L. Ludwig
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - David M. Gamm
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53705, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Comparative Biomedical Sciences, University of Wisconsin–Madison, Madison, WI, 53706, USA
| | - Shaoqin Gong
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Zhenqiang Ma
- Department of Electrical and Computer Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Grainger Institute for Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| |
Collapse
|
40
|
Bohrer LR, Stone NE, Wright AT, Han S, Sicher I, Sulchek TA, Mullins RF, Tucker BA. CGMP Compliant Microfluidic Transfection of Induced Pluripotent Stem Cells for CRISPR-Mediated Genome Editing. Stem Cells 2023; 41:1037-1046. [PMID: 37632456 PMCID: PMC10631803 DOI: 10.1093/stmcls/sxad063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 07/26/2023] [Indexed: 08/28/2023]
Abstract
Inherited retinal degeneration is a term used to describe heritable disorders that result from the death of light sensing photoreceptor cells. Although we and others believe that it will be possible to use gene therapy to halt disease progression early in its course, photoreceptor cell replacement will likely be required for patients who have already lost their sight. While advances in autologous photoreceptor cell manufacturing have been encouraging, development of technologies capable of efficiently delivering genome editing reagents to stem cells using current good manufacturing practices (cGMP) are needed. Gene editing reagents were delivered to induced pluripotent stem cells (iPSCs) using a Zephyr microfluidic transfection platform (CellFE). CRISPR-mediated cutting was quantified using an endonuclease assay. CRISPR correction was confirmed via digital PCR and Sanger sequencing. The resulting corrected cells were also karyotyped and differentiated into retinal organoids. We describe use of a novel microfluidic transfection platform to correct, via CRISPR-mediated homology-dependent repair (HDR), a disease-causing NR2E3 mutation in patient-derived iPSCs using cGMP compatible reagents and approaches. We show that the resulting cell lines have a corrected genotype, exhibit no off-target cutting, retain pluripotency and a normal karyotype and can be differentiated into retinal tissue suitable for transplantation. The ability to codeliver CRISPR/Cas9 and HDR templates to patient-derived iPSCs without using proprietary transfection reagents will streamline manufacturing protocols, increase the safety of resulting cell therapies, and greatly reduce the regulatory burden of clinical trials.
Collapse
Affiliation(s)
- Laura R Bohrer
- Institute for Vision Research, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Nicholas E Stone
- Institute for Vision Research, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Allison T Wright
- Institute for Vision Research, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | | | | | - Todd A Sulchek
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Robert F Mullins
- Institute for Vision Research, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Budd A Tucker
- Institute for Vision Research, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
41
|
Kruczek K, Swaroop A. Patient stem cell-derived in vitro disease models for developing novel therapies of retinal ciliopathies. Curr Top Dev Biol 2023; 155:127-163. [PMID: 38043950 DOI: 10.1016/bs.ctdb.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Primary cilia are specialized organelles on the surface of almost all cells in vertebrate tissues and are primarily involved in the detection of extracellular stimuli. In retinal photoreceptors, cilia are uniquely modified to form outer segments containing components required for the detection of light in stacks of membrane discs. Not surprisingly, vision impairment is a frequent phenotype associated with ciliopathies, a heterogeneous class of conditions caused by mutations in proteins required for formation, maintenance and/or function of primary cilia. Traditionally, immortalized cell lines and model organisms have been used to provide insights into the biology of ciliopathies. The advent of methods for reprogramming human somatic cells into pluripotent stem cells has enabled the generation of in vitro disease models directly from patients suffering from ciliopathies. Such models help us in investigating pathological mechanisms specific to human physiology and in developing novel therapeutic approaches. In this article, we review current protocols to differentiate human pluripotent stem cells into retinal cell types, and discuss how these cellular and/or organoid models can be utilized to interrogate pathobiology of ciliopathies affecting the retina and for testing prospective treatments.
Collapse
Affiliation(s)
- Kamil Kruczek
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD, United States.
| | - Anand Swaroop
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
42
|
Gravina A, Tediashvili G, Zheng Y, Iwabuchi KA, Peyrot SM, Roodsari SZ, Gargiulo L, Kaneko S, Osawa M, Schrepfer S, Deuse T. Synthetic immune checkpoint engagers protect HLA-deficient iPSCs and derivatives from innate immune cell cytotoxicity. Cell Stem Cell 2023; 30:1538-1548.e4. [PMID: 37922880 DOI: 10.1016/j.stem.2023.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 08/23/2023] [Accepted: 10/04/2023] [Indexed: 11/07/2023]
Abstract
Immune rejection of allogeneic cell therapeutics remains a major problem for immuno-oncology and regenerative medicine. Allogeneic cell products so far have inferior persistence and efficacy when compared with autologous alternatives. Engineering of hypoimmune cells may greatly improve their therapeutic benefit. We present a new class of agonistic immune checkpoint engagers that protect human leukocyte antigen (HLA)-depleted induced pluripotent stem cell-derived endothelial cells (iECs) from innate immune cells. Engagers with agonistic functionality to their inhibitory receptors TIM3 and SIRPα effectively protect engineered iECs from natural killer (NK) cell and macrophage killing. The SIRPα engager can be combined with truncated CD64 to generate fully immune evasive iECs capable of escaping allogeneic cellular and immunoglobulin G (IgG) antibody-mediated rejection. Synthetic immune checkpoint engagers have high target specificity and lack retrograde signaling in the engineered cells. This modular design allows for the exploitation of more inhibitory immune pathways for immune evasion and could contribute to the advancement of allogeneic cell therapeutics.
Collapse
Affiliation(s)
- Alessia Gravina
- Transplant and Stem Cell Immunobiology (TSI)-Lab, Department of Surgery, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Grigol Tediashvili
- Transplant and Stem Cell Immunobiology (TSI)-Lab, Department of Surgery, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Yueting Zheng
- Shinobi Therapeutics, 2 Tower Place, South San Francisco, CA 94080, USA
| | - Kumiko A Iwabuchi
- Shinobi Therapeutics, 2 Tower Place, South San Francisco, CA 94080, USA
| | - Sara M Peyrot
- Shinobi Therapeutics, 2 Tower Place, South San Francisco, CA 94080, USA
| | - Susan Z Roodsari
- Shinobi Therapeutics, 2 Tower Place, South San Francisco, CA 94080, USA
| | - Lauren Gargiulo
- Shinobi Therapeutics, 2 Tower Place, South San Francisco, CA 94080, USA
| | - Shin Kaneko
- Laboratory of Regenerative Immunotherapy, Department of Cell Growth and Differentiation, Center for iPS cell Research, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Mitsujiro Osawa
- Shinobi Therapeutics, Med-Pharm Collaboration Building 46-29, Yoshida-Shimo-Adachi-Cho, Sakyo-Ku, Kyoto, Japan
| | - Sonja Schrepfer
- Transplant and Stem Cell Immunobiology (TSI)-Lab, Department of Surgery, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Tobias Deuse
- Transplant and Stem Cell Immunobiology (TSI)-Lab, Department of Surgery, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA.
| |
Collapse
|
43
|
Li J, Zeng Q. Trim9 regulates the directional differentiation of retinal Müller cells to retinal ganglion cells. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2023; 48:1561-1571. [PMID: 38432885 PMCID: PMC10929896 DOI: 10.11817/j.issn.1672-7347.2023.230108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Indexed: 03/05/2024]
Abstract
OBJECTIVES Glaucoma is a leading cause of irreversible blindness, and effective therapies to reverse the visual system damage caused by glaucoma are still lacking. Recently, the stem cell therapy enable the repair and regeneration of the damaged retinal neurons, but challenges regarding the source of stem cells remain. This study aims to investigate a protocol that allows the dedifferentiation of Müller cells into retinal stem cells, following by directed differentiation into retinal ganglion cells with high efficiency, and to provide a new method of cellular acquisition for retinal stem cells. METHODS Epidermal cell growth factor and fibroblast growth factor 2 were used to induce the dedifferentiation of rat retinal Müller cells into retinal neural stem cells. Retinal stem cells derived from Müller cells were infected with a Trim9 overexpression lentiviral vector (PGC-FU-Trim9-GFP), and the efficiency of viral infection was assessed by fluorescence microscopy and flow cytometry. Retinoic acid and brain-derived neurotrophic factor treatments were used to induce the differentiation of the retinal stem cells into neurons and glial cells with or without the overexpression of Trim9. The expressions of each cellular marker (GLAST, GS, rhodopsin, PKC, HPC-1, Calbindin, Thy1.1, Brn-3b, Nestin, Pax6) were detected by immunofluorescence, PCR/real-time RT-PCR or Western blotting. RESULTS Rat retinal Müller cells expressed neural stem cells markers (Nestin and Pax6) with the treatment of epidermal cell growth factor and fibroblast growth factor 2. The Thy1.1 positive cell rate of retinal stem cells overexpressing Trim9 was significantly increased, indicating their directional differentiation into retinal ganglion cells after treatment with retinoic acid and brain-derived neurotrophic factor. CONCLUSIONS In this study, rat retinal Müller cells are dedifferentiated into retinal stem cells successfully, and Trim9 promotes the directional differentiation from retinal stem cells to retinal ganglion cells effectively.
Collapse
Affiliation(s)
- Jinxiang Li
- Department of Ophthalmology, First Hospital Affiliated with Hunan Normal University (Hunan Provincial People's Hospital), Changsha 410005, China.
| | - Qi Zeng
- Department of Ophthalmology, First Hospital Affiliated with Hunan Normal University (Hunan Provincial People's Hospital), Changsha 410005, China.
| |
Collapse
|
44
|
Kabra M, Shahi PK, Wang Y, Sinha D, Spillane A, Newby GA, Saxena S, Tong Y, Chang Y, Abdeen AA, Edwards KL, Theisen CO, Liu DR, Gamm DM, Gong S, Saha K, Pattnaik BR. Nonviral base editing of KCNJ13 mutation preserves vision in a model of inherited retinal channelopathy. J Clin Invest 2023; 133:e171356. [PMID: 37561581 PMCID: PMC10541187 DOI: 10.1172/jci171356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 08/08/2023] [Indexed: 08/12/2023] Open
Abstract
Clinical genome editing is emerging for rare disease treatment, but one of the major limitations is the targeting of CRISPR editors' delivery. We delivered base editors to the retinal pigmented epithelium (RPE) in the mouse eye using silica nanocapsules (SNCs) as a treatment for retinal degeneration. Leber congenital amaurosis type 16 (LCA16) is a rare pediatric blindness caused by point mutations in the KCNJ13 gene, a loss of function inwardly rectifying potassium channel (Kir7.1) in the RPE. SNCs carrying adenine base editor 8e (ABE8e) mRNA and sgRNA precisely and efficiently corrected the KCNJ13W53X/W53X mutation. Editing in both patient fibroblasts (47%) and human induced pluripotent stem cell-derived RPE (LCA16-iPSC-RPE) (17%) showed minimal off-target editing. We detected functional Kir7.1 channels in the edited LCA16-iPSC-RPE. In the LCA16 mouse model (Kcnj13W53X/+ΔR), RPE cells targeted SNC delivery of ABE8e mRNA preserved normal vision, measured by full-field electroretinogram (ERG). Moreover, multifocal ERG confirmed the topographic measure of electrical activity primarily originating from the edited retinal area at the injection site. Preserved retina structure after treatment was established by optical coherence tomography (OCT). This preclinical validation of targeted ion channel functional rescue, a challenge for pharmacological and genomic interventions, reinforced the effectiveness of nonviral genome-editing therapy for rare inherited disorders.
Collapse
Affiliation(s)
- Meha Kabra
- Department of Pediatrics
- McPherson Eye Research Institute
| | - Pawan K. Shahi
- Department of Pediatrics
- McPherson Eye Research Institute
| | - Yuyuan Wang
- Department of Biomedical Engineering
- Wisconsin Institute of Discovery, and
| | - Divya Sinha
- McPherson Eye Research Institute
- Waisman Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | | | - Gregory A. Newby
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Howard Hughes Medical Institute and
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Shivani Saxena
- McPherson Eye Research Institute
- Department of Biomedical Engineering
- Wisconsin Institute of Discovery, and
| | - Yao Tong
- Department of Biomedical Engineering
- Wisconsin Institute of Discovery, and
| | | | - Amr A. Abdeen
- McPherson Eye Research Institute
- Department of Biomedical Engineering
- Wisconsin Institute of Discovery, and
| | - Kimberly L. Edwards
- McPherson Eye Research Institute
- Waisman Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Cole O. Theisen
- Waisman Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - David R. Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Howard Hughes Medical Institute and
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA
| | - David M. Gamm
- McPherson Eye Research Institute
- Waisman Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
- Department of Ophthalmology and Visual Sciences and
| | - Shaoqin Gong
- McPherson Eye Research Institute
- Department of Biomedical Engineering
- Wisconsin Institute of Discovery, and
- Department of Ophthalmology and Visual Sciences and
| | - Krishanu Saha
- Department of Pediatrics
- McPherson Eye Research Institute
- Department of Biomedical Engineering
- Wisconsin Institute of Discovery, and
- Center for Human Genomics and Precision Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Bikash R. Pattnaik
- Department of Pediatrics
- McPherson Eye Research Institute
- Department of Ophthalmology and Visual Sciences and
| |
Collapse
|
45
|
Lapshin EV, Gershovich YG, Karabelsky AV. The Potential and Application of iPSCs in Gene and Cell Therapy for Retinopathies and Optic Neuropathies. Acta Naturae 2023; 15:56-64. [PMID: 38234607 PMCID: PMC10790360 DOI: 10.32607/actanaturae.25454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 10/05/2023] [Indexed: 01/19/2024] Open
Abstract
This review focuses on in vitro modeling of diseases and the development of therapeutic strategies using iPSCs for the two most common types of optical pathologies: hereditary neuropathies and retinopathies. Degeneration of retinal ganglion cells and the subsequent optic nerve atrophy leads to various types of neuropathies. Damage to photoreceptor cells or retinal pigment epithelium cells causes various retinopathies. Human iPSCs can be used as a model for studying the pathological foundations of diseases and for developing therapies to restore visual function. In recent years, significant progress has also been made in creating ganglionic and retinal organoids from iPSCs. Different research groups have published data pertaining to the potential of using iPSCs for the modeling of optic neuropathies such as glaucoma, Leber hereditary optic neuropathy, etc., including in the development of therapeutic approaches using gene editing tools.
Collapse
Affiliation(s)
- E. V. Lapshin
- Gene Therapy Department, Science Center for Translational Medicine, Sirius University of Science and Technology, Krasnodar Region, Sirius, 354340 Russian Federation
| | - Y. G. Gershovich
- Gene Therapy Department, Science Center for Translational Medicine, Sirius University of Science and Technology, Krasnodar Region, Sirius, 354340 Russian Federation
| | - A. V. Karabelsky
- Gene Therapy Department, Science Center for Translational Medicine, Sirius University of Science and Technology, Krasnodar Region, Sirius, 354340 Russian Federation
| |
Collapse
|
46
|
Sasseville S, Karami S, Tchatchouang A, Charpentier P, Anney P, Gobert D, Proulx S. Biomaterials used for tissue engineering of barrier-forming cell monolayers in the eye. Front Bioeng Biotechnol 2023; 11:1269385. [PMID: 37840667 PMCID: PMC10569698 DOI: 10.3389/fbioe.2023.1269385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
Cell monolayers that form a barrier between two structures play an important role for the maintenance of tissue functionality. In the anterior portion of the eye, the corneal endothelium forms a barrier that controls fluid exchange between the aqueous humor of the anterior chamber and the corneal stroma. This monolayer is central in the pathogenesis of Fuchs endothelial corneal dystrophy (FECD). FECD is a common corneal disease, in which corneal endothelial cells deposit extracellular matrix that increases the thickness of its basal membrane (Descemet's membrane), and forms excrescences (guttae). With time, there is a decrease in endothelial cell density that generates vision loss. Transplantation of a monolayer of healthy corneal endothelial cells on a Descemet membrane substitute could become an interesting alternative for the treatment of this pathology. In the back of the eye, the retinal pigment epithelium (RPE) forms the blood-retinal barrier, controlling fluid exchange between the choriocapillaris and the photoreceptors of the outer retina. In the retinal disease dry age-related macular degeneration (dry AMD), deposits (drusen) form between the RPE and its basal membrane (Bruch's membrane). These deposits hinder fluid exchange, resulting in progressive RPE cell death, which in turn generates photoreceptor cell death, and vision loss. Transplantation of a RPE monolayer on a Bruch's membrane/choroidal stromal substitute to replace the RPE before photoreceptor cell death could become a treatment alternative for this eye disease. This review will present the different biomaterials that are proposed for the engineering of a monolayer of corneal endothelium for the treatment of FECD, and a RPE monolayer for the treatment of dry AMD.
Collapse
Affiliation(s)
- Samantha Sasseville
- Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX; Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC, Canada
- Département d’ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Samira Karami
- Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX; Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC, Canada
- Département d’ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Ange Tchatchouang
- Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX; Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC, Canada
- Département d’ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Pascale Charpentier
- Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX; Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC, Canada
- Département d’ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Princia Anney
- Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX; Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC, Canada
- Département d’ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Delphine Gobert
- Département d’ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de Médecine, Université Laval, Québec, QC, Canada
- Centre universitaire d’ophtalmologie (CUO), Hôpital du Saint-Sacrement, CHU de Québec-Université Laval, Québec, QC, Canada
| | - Stéphanie Proulx
- Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX; Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC, Canada
- Département d’ophtalmologie et d’oto-rhino-laryngologie-chirurgie cervico-faciale, Faculté de Médecine, Université Laval, Québec, QC, Canada
| |
Collapse
|
47
|
Lu B, Avalos P, Svendsen S, Zhang C, Nocito L, Jones MK, Pieplow C, Saylor J, Ghiam S, Block A, Fernandez M, Ljubimov AV, Small K, Liao D, Svendsen CN, Wang S. GMP-grade human neural progenitors delivered subretinally protect vision in rat model of retinal degeneration and survive in minipigs. J Transl Med 2023; 21:650. [PMID: 37743503 PMCID: PMC10519102 DOI: 10.1186/s12967-023-04501-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 09/02/2023] [Indexed: 09/26/2023] Open
Abstract
BACKGROUND Stem cell products are increasingly entering early stage clinical trials for treating retinal degeneration. The field is learning from experience about comparability of cells proposed for preclinical and clinical use. Without this, preclinical data supporting translation to a clinical study might not adequately reflect the performance of subsequent clinical-grade cells in patients. METHODS Research-grade human neural progenitor cells (hNPC) and clinical-grade hNPC (termed CNS10-NPC) were injected into the subretinal space of the Royal College of Surgeons (RCS) rat, a rodent model of retinal degeneration such as retinitis pigmentosa. An investigational new drug (IND)-enabling study with CNS10-NPC was performed in the same rodent model. Finally, surgical methodology for subretinal cell delivery in the clinic was optimized in a large animal model with Yucatan minipigs. RESULTS Both research-grade hNPC and clinical-grade hNPC can survive and provide functional and morphological protection in a dose-dependent fashion in RCS rats and the optimal cell dose was defined and used in IND-enabling studies. Grafted CNS10-NPC migrated from the injection site without differentiation into retinal cell phenotypes. Additionally, CNS10-NPC showed long-term survival, safety and efficacy in a good laboratory practice (GLP) toxicity and tumorigenicity study, with no observed cell overgrowth even at the maximum deliverable dose. Finally, using a large animal model with the Yucatan minipig, which has an eye size comparable to the human, we optimized the surgical methodology for subretinal cell delivery in the clinic. CONCLUSIONS These extensive studies supported an approved IND and the translation of CNS10-NPC to an ongoing Phase 1/2a clinical trial (NCT04284293) for the treatment of retinitis pigmentosa.
Collapse
Affiliation(s)
- Bin Lu
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Pablo Avalos
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Soshana Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Changqing Zhang
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Laura Nocito
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Melissa K Jones
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Cosmo Pieplow
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Joshua Saylor
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Sean Ghiam
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Amanda Block
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Michael Fernandez
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Alexander V Ljubimov
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Kent Small
- Macula& Retina Institute, Glendale, CA, 91203, USA
| | - David Liao
- Retina Vitreous Associates Medical Group, Beverly Hills, CA, 90211, USA
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| | - Shaomei Wang
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
48
|
Wang B, Starr AL, Fraser HB. Cell type-specific cis-regulatory divergence in gene expression and chromatin accessibility revealed by human-chimpanzee hybrid cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.22.541747. [PMID: 37292820 PMCID: PMC10245923 DOI: 10.1101/2023.05.22.541747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Although gene expression divergence has long been postulated to be the primary driver of human evolution, identifying the genes and genetic variants underlying uniquely human traits has proven to be quite challenging. Theory suggests that cell type-specific cis-regulatory variants may fuel evolutionary adaptation due to the specificity of their effects. These variants can precisely tune the expression of a single gene in a single cell type, avoiding the potentially deleterious consequences of trans-acting changes and non-cell type-specific changes that can impact many genes and cell types, respectively. It has recently become possible to quantify human-specific cis-acting regulatory divergence by measuring allele-specific expression in human-chimpanzee hybrid cells-the product of fusing induced pluripotent stem (iPS) cells of each species in vitro. However, these cis-regulatory changes have only been explored in a limited number of cell types. Here, we quantify human-chimpanzee cis-regulatory divergence in gene expression and chromatin accessibility across six cell types, enabling the identification of highly cell type-specific cis-regulatory changes. We find that cell type-specific genes and regulatory elements evolve faster than those shared across cell types, suggesting an important role for genes with cell type-specific expression in human evolution. Furthermore, we identify several instances of lineage-specific natural selection that may have played key roles in specific cell types, such as coordinated changes in the cis-regulation of dozens of genes involved in neuronal firing in motor neurons. Finally, using novel metrics and a machine learning model, we identify genetic variants that likely alter chromatin accessibility and transcription factor binding, leading to neuron-specific changes in the expression of the neurodevelopmentally important genes FABP7 and GAD1. Overall, our results demonstrate that integrative analysis of cis-regulatory divergence in chromatin accessibility and gene expression across cell types is a promising approach to identify the specific genes and genetic variants that make us human.
Collapse
Affiliation(s)
- Ban Wang
- Department of Biology, Stanford University, Stanford, CA, USA
| | | | | |
Collapse
|
49
|
Patel SH, Lamba DA. Factors Affecting Stem Cell-Based Regenerative Approaches in Retinal Degeneration. Annu Rev Vis Sci 2023; 9:155-175. [PMID: 37713278 DOI: 10.1146/annurev-vision-120222-012817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Inherited and age-associated vision loss is often associated with degeneration of the cells of the retina, the light-sensitive layer at the back of the eye. The mammalian retina, being a postmitotic neural tissue, does not have the capacity to repair itself through endogenous regeneration. There has been considerable excitement for the development of cell replacement approaches since the isolation and development of culture methods for human pluripotent stem cells, as well as the generation of induced pluripotent stem cells. This has now been combined with novel three-dimensional organoid culture systems that closely mimic human retinal development in vitro. In this review, we cover the current state of the field, with emphasis on the cell delivery challenges, role of the recipient immunological microenvironment, and challenges related to connectivity between transplanted cells and host circuitry both locally and centrally to the different areas of the brain.
Collapse
Affiliation(s)
- Sachin H Patel
- Department of Ophthalmology, University of California, San Francisco, California, USA;
| | - Deepak A Lamba
- Department of Ophthalmology, University of California, San Francisco, California, USA;
- Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, University of California, San Francisco, California, USA
| |
Collapse
|
50
|
Wei X, Wang L, Duan C, Chen K, Li X, Guo X, Chen P, Liu H, Fan Y. Cardiac patches made of brown adipose-derived stem cell sheets and conductive electrospun nanofibers restore infarcted heart for ischemic myocardial infarction. Bioact Mater 2023; 27:271-287. [PMID: 37122901 PMCID: PMC10130885 DOI: 10.1016/j.bioactmat.2023.03.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 03/26/2023] [Accepted: 03/30/2023] [Indexed: 05/02/2023] Open
Abstract
Cell sheet engineering has been proven to be a promising strategy for cardiac remodeling post-myocardial infarction. However, insufficient mechanical strength and low cell retention lead to limited therapeutic efficiency. The thickness and area of artificial cardiac patches also affect their therapeutic efficiency. Cardiac patches prepared by combining cell sheets with electrospun nanofibers, which can be transplanted and sutured to the surface of the infarcted heart, promise to solve this problem. Here, we fabricated a novel cardiac patch by stacking brown adipose-derived stem cells (BADSCs) sheet layer by layer, and then they were combined with multi-walled carbon nanotubes (CNTs)-containing electrospun polycaprolactone/silk fibroin nanofibers (CPSN). The results demonstrated that BADSCs tended to generate myocardium-like structures seeded on CPSN. Compared with BADSCs suspension-containing electrospun nanofibers, the transplantation of the CPSN-BADSCs sheets (CNBS) cardiac patches exhibited accelerated angiogenesis and decreased inflammation in a rat myocardial infarction model. In addition, the CNBS cardiac patches could regulate macrophage polarization and promote gap junction remodeling, thus restoring cardiac functions. Overall, the hybrid cardiac patches made of electrospun nanofibers and cell sheets provide a novel solution to cardiac remodeling after ischemic myocardial infarction.
Collapse
Affiliation(s)
- Xinbo Wei
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
| | - Li Wang
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
| | - Cuimi Duan
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences, Beijing, 100850, PR China
| | - Kai Chen
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
| | - Xia Li
- Beijing Citident Stomatology Hospital, Beijing, 100032, PR China
| | - Ximin Guo
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences, Beijing, 100850, PR China
| | - Peng Chen
- Department of Ultrasound, The Third Medical Center, Chinese PLA General Hospital, Beijing, PR China
| | - Haifeng Liu
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
| |
Collapse
|