1
|
Liu J, Zhang F, Shi X. The role of metal nanocarriers, liposomes and chitosan-based nanoparticles in diabetic retinopathy treatment: A review study. Int J Biol Macromol 2025; 291:139017. [PMID: 39708854 DOI: 10.1016/j.ijbiomac.2024.139017] [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: 10/04/2024] [Revised: 12/04/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
Diabetic Retinopathy (DR) is a significant and progressive eye complication associated with diabetes mellitus, leading to potential vision loss. The pathophysiology of DR involves complex neurovascular changes due to prolonged hyperglycemia, resulting in microangiopathy and neurodegeneration. Current treatment modalities come with limitations such as low bioavailability of therapeutic agents, risk of side effects, and surgical complications. Consequently, the prevention and management of DR, particularly in its advanced stages, present ongoing challenges. This review investigates recent advancements in nanotechnology as a novel approach to enhance the treatment of DR. A comprehensive literature review of recent studies focusing on nanocarriers for drug delivery in DR treatment and an analysis of their efficacy compared to traditional methods was conducted for this study. The findings indicate that nanotechnology can significantly enhance the bioavailability of therapeutic agents while minimizing systemic exposure and associated side effects. The novelty of this study lies in its focus on the intersection of nanotechnology and ophthalmology, exploring innovative solutions that extend beyond existing literature on DR treatments. By highlighting recent advancements in this field, the study paves the way for future research aimed at developing more effective therapeutic strategies for managing DR.
Collapse
Affiliation(s)
- Junling Liu
- Linqu Zhengda Guangming Eye Hospital, Zhengda Guangming Eye Group, Weifang 262600, Shandong, China
| | - Feng Zhang
- Linqu Zhengda Guangming Eye Hospital, Zhengda Guangming Eye Group, Weifang 262600, Shandong, China.
| | - Xiaolong Shi
- Linqu Zhengda Guangming Eye Hospital, Zhengda Guangming Eye Group, Weifang 262600, Shandong, China
| |
Collapse
|
2
|
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
|
3
|
Ning K, Bhuckory MB, Lo CH, Sendayen BE, Kowal TJ, Chen M, Bansal R, Chang KC, Vollrath D, Berbari NF, Mahajan VB, Hu Y, Sun Y. Cilia-associated wound repair mediated by IFT88 in retinal pigment epithelium. Sci Rep 2023; 13:8205. [PMID: 37211572 PMCID: PMC10200793 DOI: 10.1038/s41598-023-35099-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/12/2023] [Indexed: 05/23/2023] Open
Abstract
Primary cilia are conserved organelles that integrate extracellular cues into intracellular signals and are critical for diverse processes, including cellular development and repair responses. Deficits in ciliary function cause multisystemic human diseases known as ciliopathies. In the eye, atrophy of the retinal pigment epithelium (RPE) is a common feature of many ciliopathies. However, the roles of RPE cilia in vivo remain poorly understood. In this study, we first found that mouse RPE cells only transiently form primary cilia. We then examined the RPE in the mouse model of Bardet-Biedl Syndrome 4 (BBS4), a ciliopathy associated with retinal degeneration in humans, and found that ciliation in BBS4 mutant RPE cells is disrupted early during development. Next, using a laser-induced injury model in vivo, we found that primary cilia in RPE reassemble in response to laser injury during RPE wound healing and then rapidly disassemble after the repair is completed. Finally, we demonstrated that RPE-specific depletion of primary cilia in a conditional mouse model of cilia loss promoted wound healing and enhanced cell proliferation. In summary, our data suggest that RPE cilia contribute to both retinal development and repair and provide insights into potential therapeutic targets for more common RPE degenerative diseases.
Collapse
Affiliation(s)
- Ke Ning
- Department of Ophthalmology, Stanford University School of Medicine, 1651 Page Mill Road, Rm 2220, Palo Alto, CA, 94304, USA
| | - Mohajeet B Bhuckory
- Department of Ophthalmology, Stanford University School of Medicine, 1651 Page Mill Road, Rm 2220, Palo Alto, CA, 94304, USA
| | - Chien-Hui Lo
- Department of Ophthalmology, Stanford University School of Medicine, 1651 Page Mill Road, Rm 2220, Palo Alto, CA, 94304, USA
| | - Brent E Sendayen
- Department of Ophthalmology, Stanford University School of Medicine, 1651 Page Mill Road, Rm 2220, Palo Alto, CA, 94304, USA
- Palo Alto Veterans Administration, Palo Alto, CA, USA
| | - Tia J Kowal
- Department of Ophthalmology, Stanford University School of Medicine, 1651 Page Mill Road, Rm 2220, Palo Alto, CA, 94304, USA
| | - Ming Chen
- Department of Ophthalmology, Stanford University School of Medicine, 1651 Page Mill Road, Rm 2220, Palo Alto, CA, 94304, USA
| | - Ruchi Bansal
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Kun-Che Chang
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Douglas Vollrath
- Department of Ophthalmology, Stanford University School of Medicine, 1651 Page Mill Road, Rm 2220, Palo Alto, CA, 94304, USA
- Department of Genetics, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Nicolas F Berbari
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Vinit B Mahajan
- Department of Ophthalmology, Stanford University School of Medicine, 1651 Page Mill Road, Rm 2220, Palo Alto, CA, 94304, USA
| | - Yang Hu
- Department of Ophthalmology, Stanford University School of Medicine, 1651 Page Mill Road, Rm 2220, Palo Alto, CA, 94304, USA
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, 1651 Page Mill Road, Rm 2220, Palo Alto, CA, 94304, USA.
- Palo Alto Veterans Administration, Palo Alto, CA, USA.
| |
Collapse
|
4
|
Cohen-Gulkar M, David A, Messika-Gold N, Eshel M, Ovadia S, Zuk-Bar N, Idelson M, Cohen-Tayar Y, Reubinoff B, Ziv T, Shamay M, Elkon R, Ashery-Padan R. The LHX2-OTX2 transcriptional regulatory module controls retinal pigmented epithelium differentiation and underlies genetic risk for age-related macular degeneration. PLoS Biol 2023; 21:e3001924. [PMID: 36649236 PMCID: PMC9844853 DOI: 10.1371/journal.pbio.3001924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 11/16/2022] [Indexed: 01/18/2023] Open
Abstract
Tissue-specific transcription factors (TFs) control the transcriptome through an association with noncoding regulatory regions (cistromes). Identifying the combination of TFs that dictate specific cell fate, their specific cistromes and examining their involvement in complex human traits remain a major challenge. Here, we focus on the retinal pigmented epithelium (RPE), an essential lineage for retinal development and function and the primary tissue affected in age-related macular degeneration (AMD), a leading cause of blindness. By combining mechanistic findings in stem-cell-derived human RPE, in vivo functional studies in mice and global transcriptomic and proteomic analyses, we revealed that the key developmental TFs LHX2 and OTX2 function together in transcriptional module containing LDB1 and SWI/SNF (BAF) to regulate the RPE transcriptome. Importantly, the intersection between the identified LHX2-OTX2 cistrome with published expression quantitative trait loci, ATAC-seq data from human RPE, and AMD genome-wide association study (GWAS) data, followed by functional validation using a reporter assay, revealed a causal genetic variant that affects AMD risk by altering TRPM1 expression in the RPE through modulation of LHX2 transcriptional activity on its promoter. Taken together, the reported cistrome of LHX2 and OTX2, the identified downstream genes and interacting co-factors reveal the RPE transcription module and uncover a causal regulatory risk single-nucleotide polymorphism (SNP) in the multifactorial common blinding disease AMD.
Collapse
Affiliation(s)
- Mazal Cohen-Gulkar
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine and Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Ahuvit David
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine and Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Naama Messika-Gold
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine and Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Mai Eshel
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine and Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Shai Ovadia
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine and Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Nitay Zuk-Bar
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine and Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Maria Idelson
- The Hadassah Human Embryonic Stem Cell Research Center, The Goldyne Savad Institute of Gene Therapy and Department of Gynecology, Jerusalem, Israel
| | - Yamit Cohen-Tayar
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine and Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Benjamin Reubinoff
- The Hadassah Human Embryonic Stem Cell Research Center, The Goldyne Savad Institute of Gene Therapy and Department of Gynecology, Jerusalem, Israel
| | - Tamar Ziv
- Smoler Proteomics Center, Lorry I. Lokey Interdisciplinary Center for Life Sciences and Engineering, Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Meir Shamay
- Daniella Lee Casper Laboratory in Viral Oncology, Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Ran Elkon
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine and Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
- * E-mail: (RE); (RAP)
| | - Ruth Ashery-Padan
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine and Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
- * E-mail: (RE); (RAP)
| |
Collapse
|
5
|
Gusev AA, Zakharova OV, Vasyukova IA, Osmanov RE, Al-Makhdar YM. [Nanotechnologies in ophthalmology]. Vestn Oftalmol 2023; 139:107-114. [PMID: 37638580 DOI: 10.17116/oftalma2023139041107] [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: 08/29/2023]
Abstract
Application of new materials and methods in the diagnosis and treatment of eye diseases is one of the promising research areas in modern ophthalmology. Significant progress has been made in understanding the pathogenesis, diagnosis and treatment of eye diseases using nanotechnologies and nanomaterials. This paper presents the main achievements and results of original research on this issue. It has been shown that nanoparticles are able to overcome biological barriers, deliver drugs to the target site, and provide the required drug release rate. Modern nanotechnological approaches in tissue engineering are also being actively introduced into ophthalmology, making it possible to create nanoframeworks for growing three-dimensional cellular structures, including arrays of pigment epithelium cells and retinal ganglion cells for the treatment of retinal damage caused by degenerative diseases, injuries and infections.
Collapse
Affiliation(s)
- A A Gusev
- Tambov State University named after G.R. Derzhavin, Tambov, Russia
- National University of Science and Technology (MISIS), Moscow, Russia
| | - O V Zakharova
- Tambov State University named after G.R. Derzhavin, Tambov, Russia
- National University of Science and Technology (MISIS), Moscow, Russia
- Plekhanov Russian University of Economics, Moscow, Russia
| | - I A Vasyukova
- Tambov State University named after G.R. Derzhavin, Tambov, Russia
| | - R E Osmanov
- Tambov branch of S.N. Fedorov National Medical Research Center "MNTK "Eye Microsurgery", Tambov, Russia
| | | |
Collapse
|
6
|
Rizzolo LJ, Nasonkin IO, Adelman RA. Retinal Cell Transplantation, Biomaterials, and In Vitro Models for Developing Next-generation Therapies of Age-related Macular Degeneration. Stem Cells Transl Med 2022; 11:269-281. [PMID: 35356975 PMCID: PMC8968686 DOI: 10.1093/stcltm/szac001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 12/02/2021] [Indexed: 11/12/2022] Open
Abstract
Retinal pigment epithelium (RPE) cells grown on a scaffold, an RPE patch, have potential to ameliorate visual impairment in a limited number of retinal degenerative conditions. This tissue-replacement therapy is suited for age-related macular degeneration (AMD), and related diseases. RPE cells must be transplanted before the disease reaches a point of no return, represented by the loss of photoreceptors. Photoreceptors are specialized, terminally differentiated neurosensory cells that must interact with RPE's apical processes to be functional. Human photoreceptors are not known to regenerate. On the RPE's basal side, the RPE transplant must induce the reformation of the choriocapillaris, thereby re-establishing the outer blood-retinal barrier. Because the scaffold is positioned between the RPE and choriocapillaris, it should ideally degrade and be replaced by the natural extracellular matrix that separates these tissues. Besides biodegradable, the scaffolds need to be nontoxic, thin enough to not affect the focal length of the eye, strong enough to survive the transplant procedure, yet flexible enough to conform to the curvature of the retina. The challenge is patients with progressing AMD treasure their remaining vision and fear that a risky surgical procedure will further degrade their vision. Accordingly, clinical trials only treat eyes with severe impairment that have few photoreceptors to interact with the transplanted patch. Although safety has been demonstrated, the cell-replacement mechanism and efficacy remain difficult to validate. This review covers the structure of the retina, the pathology of AMD, the limitations of cell therapy approaches, and the recent progress in developing retinal therapies using biomaterials.
Collapse
Affiliation(s)
- Lawrence J Rizzolo
- Department of Ophthalmology and Visual Science, Yale University, New Haven, CT, USA
- Department of Surgery, Yale University, New Haven, CT, USA
| | | | - Ron A Adelman
- Department of Ophthalmology and Visual Science, Yale University, New Haven, CT, USA
| |
Collapse
|
7
|
Bucher K, Rodriguez-Bocanegra E, Fischer MD. Benefits and Shortcomings of Laboratory Model Systems in the Development of Genetic Therapies. Klin Monbl Augenheilkd 2022; 239:263-269. [PMID: 35316853 DOI: 10.1055/a-1757-9879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Gene therapeutic approaches promise treatment or even a cure of diseases that were previously untreatable. Retinal gene therapies tested in clinical trials comprise a wide range of different strategies, including gene supplementation therapies, in vivo gene editing, modulation of splicing mechanisms, or the suppression of gene expression. To guarantee efficient transfer of genetic material into the respective target cells while avoiding major adverse effects, the development of genetic therapies requires appropriate in vitro model systems that allow tests of efficacy and safety of the gene therapeutic approach. In this review, we introduce various in vitro models of different levels of complexity used in the development of genetic therapies and discuss their respective benefits and shortcomings using the example of adeno-associated virus-based retinal gene therapy.
Collapse
Affiliation(s)
- Kirsten Bucher
- University Eye Hospital, University Hospital Tübingen Clinic of Ophthalmology, Tubingen, Germany.,Institute for Ophthalmic Research, University Hospital Tübingen Clinic of Ophthalmology, Tubingen, Germany
| | | | - M Dominik Fischer
- University Eye Hospital, University Hospital Tübingen Clinic of Ophthalmology, Tubingen, Germany.,Institute for Ophthalmic Research, University Hospital Tübingen Clinic of Ophthalmology, Tubingen, Germany.,Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom of Great Britain and Northern Ireland.,Department of Clinical Neurosciences, University of Oxford Nuffield Laboratory of Ophthalmology, Oxford, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
8
|
Matynia A, Wang J, Kim S, Li Y, Dimashkie A, Jiang Z, Hu J, Strom SP, Radu RA, Chen R, Gorin MB. Assessing Variant Causality and Severity Using Retinal Pigment Epithelial Cells Derived from Stargardt Disease Patients. Transl Vis Sci Technol 2022; 11:33. [PMID: 35348597 PMCID: PMC8976924 DOI: 10.1167/tvst.11.3.33] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/19/2022] [Indexed: 02/06/2023] Open
Abstract
Purpose Modern molecular genetics has revolutionized gene discovery, genetic diagnoses, and precision medicine yet many patients remain unable to benefit from these advances as disease-causing variants remain elusive for up to half of Mendelian genetic disorders. Patient-derived induced pluripotent stem (iPS) cells and transcriptomics were used to identify the fate of unsolved ABCA4 alleles in patients with Stargardt disease. Methods Multiple independent iPS lines were generated from skin biopsies of three patients with Stargardt disease harboring a single identified pathogenic ABCA4 variant. Derived retinal pigment epithelial cells (dRPE) from a normal control and patient cells were subjected to RNA-Seq on the Novaseq6000 platform, analyzed using DESeq2 with calculation of allele specific imbalance from the pathogenic or a known linked variant. Protein analysis was performed using the automated Simple Western system. Results Nine dRPE samples were generated, with transcriptome analysis on eight. Allele-specific expression indicated normal transcripts expressed from splice variants albeit at low levels, and missense transcripts expressed at near-normal levels. Corresponding protein was not easily detected. Patient phenotype correlation indicated missense variants expressed at high levels have more deleterious outcomes. Transcriptome analysis suggests mitochondrial membrane biodynamics and the unfolded protein response pathway may be relevant in Stargardt disease. Conclusions Patient-specific iPS-derived RPE cells set the stage to assess non-expressing variants in difficult-to-detect genomic regions using easily biopsied tissue. Translational Relevance This "Disease in a Dish" approach is likely to enhance the ability of patients to participate in and benefit from clinical trials while providing insights into perturbations in RPE biology.
Collapse
Affiliation(s)
- Anna Matynia
- UCLA Stein Eye Institute and Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Jun Wang
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Sangbae Kim
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Yumei Li
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Anupama Dimashkie
- Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
| | - Zhichun Jiang
- UCLA Stein Eye Institute and Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Jane Hu
- UCLA Stein Eye Institute and Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | | | - Roxana A. Radu
- UCLA Stein Eye Institute and Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Rui Chen
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Structural and Computational Biology and Molecular Biophysics Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Michael B. Gorin
- UCLA Stein Eye Institute and Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
9
|
Alcalde I, Sánchez-Fernández C, Martín C, De Pablo N, Jemni-Damer N, Guinea GV, Merayo-Lloves J, Del Olmo-Aguado S. Human Stem Cell Transplantation for Retinal Degenerative Diseases: Where Are We Now? MEDICINA (KAUNAS, LITHUANIA) 2022; 58:102. [PMID: 35056410 PMCID: PMC8781134 DOI: 10.3390/medicina58010102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/30/2021] [Accepted: 01/05/2022] [Indexed: 06/14/2023]
Abstract
Background and Objectives: Irreversible visual impairment is mainly caused by retinal degenerative diseases such as age-related macular degeneration and retinitis pigmentosa. Stem cell research has experienced rapid progress in recent years, and researchers and clinical ophthalmologists are trying to implement this promising technology to treat retinal degeneration. The objective of this systematic review is to analyze currently available data from clinical trials applying stem cells to treat human retinal diseases. Materials and Methods: We performed a systematic literature search in PubMed to identify articles related with stem cell therapies to retinal diseases published prior to September 2021. Furthermore, a systematic search in ClinicalTrials (NIH U.S. National Library of Medicine) was performed to identify clinical trials using stem cells to treat retinal diseases. A descriptive analysis of status, conditions, phases, interventions, and outcomes is presented here. Conclusions: To date, no available therapy based on stem cell transplantation is approved for use with patients. However, numerous clinical trials are currently finishing their initial phases and, in general, the outcomes related to implantation techniques and their long-term safety seem promising. In the next few years, we expect to see quantifiable results pertaining to visual function improvement.
Collapse
Affiliation(s)
- Ignacio Alcalde
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, Universidad de Oviedo, 33012 Oviedo, Spain; (C.S.-F.); (C.M.); (N.D.P.); (J.M.-L.); (S.D.O.-A.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Cristina Sánchez-Fernández
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, Universidad de Oviedo, 33012 Oviedo, Spain; (C.S.-F.); (C.M.); (N.D.P.); (J.M.-L.); (S.D.O.-A.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Carla Martín
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, Universidad de Oviedo, 33012 Oviedo, Spain; (C.S.-F.); (C.M.); (N.D.P.); (J.M.-L.); (S.D.O.-A.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Department of Functional Biology, University of Oviedo, 33006 Oviedo, Spain
| | - Nagore De Pablo
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, Universidad de Oviedo, 33012 Oviedo, Spain; (C.S.-F.); (C.M.); (N.D.P.); (J.M.-L.); (S.D.O.-A.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Nahla Jemni-Damer
- Neuro-Computing and Neuro-Robotics Research Group, Complutense University of Madrid, 28040 Madrid, Spain;
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital (IdISSC), 28040 Madrid, Spain
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid, 28223 Madrid, Spain;
| | - Gustavo V. Guinea
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid, 28223 Madrid, Spain;
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). 28040 Madrid, Spain
- Biomaterials and Regenerative Medicine Group, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Jesús Merayo-Lloves
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, Universidad de Oviedo, 33012 Oviedo, Spain; (C.S.-F.); (C.M.); (N.D.P.); (J.M.-L.); (S.D.O.-A.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Susana Del Olmo-Aguado
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, Universidad de Oviedo, 33012 Oviedo, Spain; (C.S.-F.); (C.M.); (N.D.P.); (J.M.-L.); (S.D.O.-A.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
10
|
Sanie-Jahromi F, Nowroozzadeh MH. RPE based gene and cell therapy for inherited retinal diseases: A review. Exp Eye Res 2022; 217:108961. [DOI: 10.1016/j.exer.2022.108961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 11/29/2022]
|
11
|
Gong M, Chen S, Chen Q, Zeng Y, Zhang Y. Generative Adversarial Networks in Medical Image Processing. Curr Pharm Des 2021; 27:1856-1868. [PMID: 33238866 DOI: 10.2174/1381612826666201125110710] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/14/2020] [Accepted: 10/21/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND The emergence of generative adversarial networks (GANs) has provided new technology and framework for the application of medical images. Specifically, a GAN requires little to no labeled data to obtain high-quality data that can be generated through competition between the generator and discriminator networks. Therefore, GANs are rapidly proving to be a state-of-the-art foundation, achieving enhanced performances in various medical applications. METHODS In this article, we introduce the principles of GANs and their various variants, deep convolutional GAN, conditional GAN, Wasserstein GAN, Info-GAN, boundary equilibrium GAN, and cycle-GAN. RESULTS All various GANs have found success in medical imaging tasks, including medical image enhancement, segmentation, classification, reconstruction, and synthesis. Furthermore, we summarize the data processing methods and evaluation indicators. Finally, we note the limitations of existing methods and the existing challenges that need to be addressed in this field. CONCLUSION Although GANs are in the initial stage of development in medical image processing, it will have a great prospect in the future.
Collapse
Affiliation(s)
- Meiqin Gong
- West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Siyu Chen
- School of Computer Science, Chengdu University of Information Technology, Chengdu 610225, China
| | - Qingyuan Chen
- School of Computer Science, Chengdu University of Information Technology, Chengdu 610225, China
| | - Yuanqi Zeng
- School of Computer Science, Chengdu University of Information Technology, Chengdu 610225, China
| | - Yongqing Zhang
- School of Computer Science, Chengdu University of Information Technology, Chengdu 610225, China
| |
Collapse
|
12
|
Jha BS, Farnoodian M, Bharti K. Regulatory considerations for developing a phase I investigational new drug application for autologous induced pluripotent stem cells-based therapy product. Stem Cells Transl Med 2021; 10:198-208. [PMID: 32946199 PMCID: PMC7848308 DOI: 10.1002/sctm.20-0242] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 08/04/2020] [Accepted: 08/17/2020] [Indexed: 12/11/2022] Open
Abstract
Induced pluripotent stem cells (iPSC)-based therapies have been hailed as the future of regenerative medicine because of their potential to provide treatment options for most degenerative diseases. A key promise of iPSC-based therapies is the possibility of an autologous transplant that may engraft better in the longer-term due to its compatibility with the patient's immune system. Despite over a decade of research, clinical translation of autologous iPSC-based therapies has been slow-partly due to a lacking pre-defined regulatory path. Here, we outline regulatory considerations for developing an autologous iPSC-based product and challenges associated with the clinical manufacturing of autologous iPSCs and their derivatives. These challenges include donor tissue source, reprogramming methods, heterogeneity of differentiated cells, controls for the manufacturing process, and preclinical considerations. A robust manufacturing process with appropriate quality controls and well-informed, prospectively designed preclinical studies provide a path toward successful approval of autologous iPSC-based therapies.
Collapse
Affiliation(s)
- Balendu Shekhar Jha
- Center for Cell Engineering, Department of Transfusion MedicineClinical Center, National Institutes of HealthBethesdaMarylandUSA
| | - Mitra Farnoodian
- Ocular and Stem Cell Translational Research Section, Ophthalmic Genetics and Visual Function BranchNational Eye Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Kapil Bharti
- Ocular and Stem Cell Translational Research Section, Ophthalmic Genetics and Visual Function BranchNational Eye Institute, National Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
13
|
Microfluidic and Microscale Assays to Examine Regenerative Strategies in the Neuro Retina. MICROMACHINES 2020; 11:mi11121089. [PMID: 33316971 PMCID: PMC7763644 DOI: 10.3390/mi11121089] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/03/2020] [Accepted: 12/05/2020] [Indexed: 12/15/2022]
Abstract
Bioengineering systems have transformed scientific knowledge of cellular behaviors in the nervous system (NS) and pioneered innovative, regenerative therapies to treat adult neural disorders. Microscale systems with characteristic lengths of single to hundreds of microns have examined the development and specialized behaviors of numerous neuromuscular and neurosensory components of the NS. The visual system is comprised of the eye sensory organ and its connecting pathways to the visual cortex. Significant vision loss arises from dysfunction in the retina, the photosensitive tissue at the eye posterior that achieves phototransduction of light to form images in the brain. Retinal regenerative medicine has embraced microfluidic technologies to manipulate stem-like cells for transplantation therapies, where de/differentiated cells are introduced within adult tissue to replace dysfunctional or damaged neurons. Microfluidic systems coupled with stem cell biology and biomaterials have produced exciting advances to restore vision. The current article reviews contemporary microfluidic technologies and microfluidics-enhanced bioassays, developed to interrogate cellular responses to adult retinal cues. The focus is on applications of microfluidics and microscale assays within mammalian sensory retina, or neuro retina, comprised of five types of retinal neurons (photoreceptors, horizontal, bipolar, amacrine, retinal ganglion) and one neuroglia (Müller), but excludes the non-sensory, retinal pigmented epithelium.
Collapse
|
14
|
Subramaniam MD, Iyer M, Nair AP, Venkatesan D, Mathavan S, Eruppakotte N, Kizhakkillach S, Chandran MK, Roy A, Gopalakrishnan AV, Vellingiri B. Oxidative stress and mitochondrial transfer: A new dimension towards ocular diseases. Genes Dis 2020; 9:610-637. [PMID: 35782976 PMCID: PMC9243399 DOI: 10.1016/j.gendis.2020.11.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 09/18/2020] [Accepted: 11/27/2020] [Indexed: 12/12/2022] Open
Abstract
Ocular cells like, retinal pigment epithelium (RPE) is a highly specialized pigmented monolayer of post-mitotic cells, which is located in the posterior segment of the eye between neuro sensory retina and vascular choroid. It functions as a selective barrier and nourishes retinal visual cells. As a result of high-level oxygen consumption of retinal cells, RPE cells are vulnerable to chronic oxidative stress and an increased level of reactive oxygen species (ROS) generated from mitochondria. These oxidative stress and ROS generation in retinal cells lead to RPE degeneration. Various sources including mtDNA damage could be an important factor of oxidative stress in RPE. Gene therapy and mitochondrial transfer studies are emerging fields in ocular disease research. For retinal degenerative diseases stem cell-based transplantation methods are developed from basic research to preclinical and clinical trials. Translational research contributions of gene and cell therapy would be a new strategy to prevent, treat and cure various ocular diseases. This review focuses on the effect of oxidative stress in ocular cell degeneration and recent translational researches on retinal degenerative diseases to cure blindness.
Collapse
Affiliation(s)
- Mohana Devi Subramaniam
- SN ONGC Department of Genetics and Molecular Biology, Vision Research Foundation, Chennai 600006, Tamil Nadu, India
- Corresponding author.
| | - Mahalaxmi Iyer
- SN ONGC Department of Genetics and Molecular Biology, Vision Research Foundation, Chennai 600006, Tamil Nadu, India
- Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641 043, Tamil Nadu, India
| | - Aswathy P. Nair
- SN ONGC Department of Genetics and Molecular Biology, Vision Research Foundation, Chennai 600006, Tamil Nadu, India
| | - Dhivya Venkatesan
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Sinnakaruppan Mathavan
- SN ONGC Department of Genetics and Molecular Biology, Vision Research Foundation, Chennai 600006, Tamil Nadu, India
| | - Nimmisha Eruppakotte
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Soumya Kizhakkillach
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Manoj kumar Chandran
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Ayan Roy
- Department of Biotechnology, Lovely Professional University, Punjab 144411, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore 600127, India
| | - Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
- Corresponding author. Human Molecular Cytogenetics and Stem Cell, Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India.Fax: +91 422 2422387.
| |
Collapse
|
15
|
Singh RK, Nasonkin IO. Limitations and Promise of Retinal Tissue From Human Pluripotent Stem Cells for Developing Therapies of Blindness. Front Cell Neurosci 2020; 14:179. [PMID: 33132839 PMCID: PMC7513806 DOI: 10.3389/fncel.2020.00179] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/25/2020] [Indexed: 12/13/2022] Open
Abstract
The self-formation of retinal tissue from pluripotent stem cells generated a tremendous promise for developing new therapies of retinal degenerative diseases, which previously seemed unattainable. Together with use of induced pluripotent stem cells or/and CRISPR-based recombineering the retinal organoid technology provided an avenue for developing models of human retinal degenerative diseases "in a dish" for studying the pathology, delineating the mechanisms and also establishing a platform for large-scale drug screening. At the same time, retinal organoids, highly resembling developing human fetal retinal tissue, are viewed as source of multipotential retinal progenitors, young photoreceptors and just the whole retinal tissue, which may be transplanted into the subretinal space with a goal of replacing patient's degenerated retina with a new retinal "patch." Both approaches (transplantation and modeling/drug screening) were projected when Yoshiki Sasai demonstrated the feasibility of deriving mammalian retinal tissue from pluripotent stem cells, and generated a lot of excitement. With further work and testing of both approaches in vitro and in vivo, a major implicit limitation has become apparent pretty quickly: the absence of the uniform layer of Retinal Pigment Epithelium (RPE) cells, which is normally present in mammalian retina, surrounds photoreceptor layer and develops and matures first. The RPE layer polarize into apical and basal sides during development and establish microvilli on the apical side, interacting with photoreceptors, nurturing photoreceptor outer segments and participating in the visual cycle by recycling 11-trans retinal (bleached pigment) back to 11-cis retinal. Retinal organoids, however, either do not have RPE layer or carry patches of RPE mostly on one side, thus directly exposing most photoreceptors in the developing organoids to neural medium. Recreation of the critical retinal niche between the apical RPE and photoreceptors, where many retinal disease mechanisms originate, is so far unattainable, imposes clear limitations on both modeling/drug screening and transplantation approaches and is a focus of investigation in many labs. Here we dissect different retinal degenerative diseases and analyze how and where retinal organoid technology can contribute the most to developing therapies even with a current limitation and absence of long and functional outer segments, supported by RPE.
Collapse
|
16
|
Suri R, Neupane YR, Jain GK, Kohli K. Recent theranostic paradigms for the management of Age-related macular degeneration. Eur J Pharm Sci 2020; 153:105489. [PMID: 32717428 DOI: 10.1016/j.ejps.2020.105489] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/07/2020] [Accepted: 07/23/2020] [Indexed: 12/21/2022]
Abstract
Degenerative diseases of eye like Age-related macular degeneration (AMD), that affects the central portion of the retina (macula), is one of the leading causes of blindness worldwide especially in the elderly population. It is classified mainly as wet and dry form. With expanding knowledge about the underlying pathophysiology of the disease, various treatment strategies are being employed to halt the course of the disease progression. Hitherto, there is no ideal therapy which can cure the disease completely, and targeting the posterior segment of the eye is yet another challenge. The purpose of this review is to summarize the recent advances in the management and treatment stratagems (therapies, delivery systems and diagnostic tools) pertaining to AMD viz. molecular targeting, stem cell therapy, nanotechnology and exosomes with special reference to newer technologies like artificial intelligence and 3D printing. Furthermore, the role of diet and nutritional supplements in the prevention and treatment of the disease has also been highlighted. The alarming increase in the said disorder around the globe demands exhaustive research and investigations in the treatment zone. This review thus additionally directs the attention towards the challenges and future perspectives of different treatment approaches for AMD.
Collapse
Affiliation(s)
- Reshal Suri
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Yub Raj Neupane
- Department of Pharmacy, National University of Singapore, 117559, Singapore
| | - Gaurav Kumar Jain
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Kanchan Kohli
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
17
|
Singh MS, Park SS, Albini TA, Canto-Soler MV, Klassen H, MacLaren RE, Takahashi M, Nagiel A, Schwartz SD, Bharti K. Retinal stem cell transplantation: Balancing safety and potential. Prog Retin Eye Res 2020; 75:100779. [PMID: 31494256 PMCID: PMC7056514 DOI: 10.1016/j.preteyeres.2019.100779] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 08/29/2019] [Accepted: 09/02/2019] [Indexed: 12/14/2022]
Abstract
Stem cell transplantation holds great promise as a potential treatment for currently incurable retinal degenerative diseases that cause poor vision and blindness. Recently, safety data have emerged from several Phase I/II clinical trials of retinal stem cell transplantation. These clinical trials, usually run in partnership with academic institutions, are based on sound preclinical studies and are focused on patient safety. However, reports of serious adverse events arising from cell therapy in other poorly regulated centers have now emerged in the lay and scientific press. While progress in stem cell research for blindness has been greeted with great enthusiasm by patients, scientists, doctors and industry alike, these adverse events have raised concerns about the safety of retinal stem cell transplantation and whether patients are truly protected from undue harm. The aim of this review is to summarize and appraise the safety of human retinal stem cell transplantation in the context of its potential to be developed into an effective treatment for retinal degenerative diseases.
Collapse
Affiliation(s)
- Mandeep S Singh
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| | - Susanna S Park
- Department of Ophthalmology & Vision Science, University of California-Davis Eye Center, Sacramento, CA, 95817, USA
| | - Thomas A Albini
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - M Valeria Canto-Soler
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Henry Klassen
- Gavin Herbert Eye Institute and Stem Cell Research Center, Irvine, CA, 92697, USA
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford and Oxford University Eye Hospital, NHS Foundation Trust, NIHR Biomedical Research Centre, Oxford, OX3 9DU, UK
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, Kobe, Hyogo, 650-0047, Japan
| | - Aaron Nagiel
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA; USC Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90007, USA
| | - Steven D Schwartz
- Stein Eye Institute, University of California Los Angeles Geffen School of Medicine, Los Angeles, CA, 90095, USA; Edythe and Eli Broad Stem Cell Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Kapil Bharti
- National Eye Institute, National Institutes of Health, Bethesda, MD, 90892, USA
| |
Collapse
|
18
|
Padi S, Manescu P, Schaub N, Hotaling N, Simon C, Bharti K, Bajcsy P. Comparison of Artificial Intelligence based approaches to cell function prediction. INFORMATICS IN MEDICINE UNLOCKED 2020; 18:https://doi.org/10.1016/j.imu.2019.100270. [PMID: 32864421 PMCID: PMC7450761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Predicting Retinal Pigment Epithelium (RPE) cell functions in stem cell implants using non-invasive bright field microscopy imaging is a critical task for clinical deployment of stem cell therapies. Such cell function predictions can be carried out using Artificial Intelligence (AI) based models. In this paper we used Traditional Machine Learning (TML) and Deep Learning (DL) based AI models for cell function prediction tasks. TML models depend on feature engineering and DL models perform feature engineering automatically but have higher modeling complexity. This work aims at exploring the tradeoffs between three approaches using TML and DL based models for RPE cell function prediction from microscopy images and at understanding the accuracy relationship between pixel-, cell feature-, and implant label-level accuracies of models. Among the three compared approaches to cell function prediction, the direct approach to cell function prediction from images is slightly more accurate in comparison to indirect approaches using intermediate segmentation and/or feature engineering steps. We also evaluated accuracy variations with respect to model selections (five TML models and two DL models) and model configurations (with and without transfer learning). Finally, we quantified the relationships between segmentation accuracy and the number of samples used for training a model, segmentation accuracy and cell feature error, and cell feature error and accuracy of implant labels. We concluded that for the RPE cell data set, there is a monotonic relationship between the number of training samples and image segmentation accuracy, and between segmentation accuracy and cell feature error, but there is no such a relationship between segmentation accuracy and accuracy of RPE implant labels.
Collapse
Affiliation(s)
- Sarala Padi
- ITL, National Institute of Standards & Technology,
Gaithersburg, MD, USA
| | - Petru Manescu
- ITL, National Institute of Standards & Technology,
Gaithersburg, MD, USA
| | | | | | - Carl Simon
- MML, National Institute of Standards & Technology,
Gaithersburg, MD, USA
| | | | - Peter Bajcsy
- ITL, National Institute of Standards & Technology,
Gaithersburg, MD, USA
| |
Collapse
|
19
|
Padi S, Manescu P, Schaub N, Hotaling N, Simon C, Bharti K, Bajcsy P. Comparison of Artificial Intelligence based approaches to cell function prediction. INFORMATICS IN MEDICINE UNLOCKED 2020. [DOI: 10.1016/j.imu.2019.100270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
20
|
Ben M’Barek K, Habeler W, Regent F, Monville C. Developing Cell-Based Therapies for RPE-Associated Degenerative Eye Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1186:55-97. [DOI: 10.1007/978-3-030-28471-8_3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
21
|
García Delgado AB, de la Cerda B, Alba Amador J, Valdés Sánchez ML, Fernández-Muñoz B, Relimpio López I, Rodríguez de la Rúa E, Díez Lloret A, Calado SM, Sánchez Pernaute R, Bhattacharya SS, Díaz Corrales FJ. Subretinal Transplant of Induced Pluripotent Stem Cell-Derived Retinal Pigment Epithelium on Nanostructured Fibrin-Agarose. Tissue Eng Part A 2019; 25:799-808. [DOI: 10.1089/ten.tea.2019.0007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Ana B. García Delgado
- Regeneration and Cell Therapy Department, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Seville, Spain
| | - Berta de la Cerda
- Regeneration and Cell Therapy Department, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Seville, Spain
| | - Julia Alba Amador
- Unidad de Producción y Reprogramación Celular, Iniciativa Andaluza en Terapias Avanzadas, Sevilla, Spain
| | - Maria Lourdes Valdés Sánchez
- Regeneration and Cell Therapy Department, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Seville, Spain
| | - Beatriz Fernández-Muñoz
- Unidad de Producción y Reprogramación Celular, Iniciativa Andaluza en Terapias Avanzadas, Sevilla, Spain
| | - Isabel Relimpio López
- University Hospital Virgen Macarena, Sevilla, Spain
- RETICS Oftared, Carlos III Institute of Health (Spain), Ministry of Health RD16/0008/0010, Sevilla, Spain
| | - Enrique Rodríguez de la Rúa
- University Hospital Virgen Macarena, Sevilla, Spain
- RETICS Oftared, Carlos III Institute of Health (Spain), Ministry of Health RD16/0008/0010, Sevilla, Spain
| | - Andrea Díez Lloret
- Regeneration and Cell Therapy Department, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Seville, Spain
| | - Sofia M. Calado
- Regeneration and Cell Therapy Department, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Seville, Spain
| | - Rosario Sánchez Pernaute
- Unidad de Producción y Reprogramación Celular, Iniciativa Andaluza en Terapias Avanzadas, Sevilla, Spain
| | - Shom S. Bhattacharya
- Regeneration and Cell Therapy Department, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Seville, Spain
| | - Francisco J. Díaz Corrales
- Regeneration and Cell Therapy Department, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Seville, Spain
| |
Collapse
|
22
|
Galloway CA, Dalvi S, Shadforth AMA, Suzuki S, Wilson M, Kuai D, Hashim A, MacDonald LA, Gamm DM, Harkin DG, Singh R. Characterization of Human iPSC-RPE on a Prosthetic Bruch's Membrane Manufactured From Silk Fibroin. Invest Ophthalmol Vis Sci 2019; 59:2792-2800. [PMID: 30025113 PMCID: PMC5989661 DOI: 10.1167/iovs.17-23157] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Purpose RPE cell transplantation as a potential treatment for AMD has been extensively investigated; however, in AMD, ultrastructural damage affects both the RPE and its underlying matrix support, the Bruch's membrane (BrM). An RPE monolayer supported by a surrogate scaffold could thus provide a more effective approach to cell-based therapy for AMD. Toward this goal, we aimed to establish a functional human induced pluripotent stem cell-derived (hiPSC)-RPE monolayer on a Bombyx mori silk fibroin (BMSF) scaffold. Methods RPE differentiated from five distinct hiPSC lines were cultured on BMSF membrane coated with extracellular matrix (ECM, COL1), and either regular tissue culture plastic or Transwell coated with ECM (LAM-TCP). Morphologic, gene and protein expression, and functional characteristics of the hiPSC-RPE cultured on different membranes were compared in longitudinal experiments spanning 1 day to ≥3 months. Results The hiPSC-RPE monolayers on ECM-coated BMSF and TCP could be maintained in culture for ≥3 months and displayed RPE-characteristic morphology, pigmentation, polarity, and expression of RPE signature genes and proteins. Furthermore, hiPSC-RPE on both ECM-coated BMSF and TCP displayed robust expression and secretion of several basement membrane proteins. Importantly, hiPSC-RPE cells on COL1-BMSF and LAM-TCP showed similar efficacy in the phagocytosis and degradation of photoreceptor outer segments. Conclusions A biomaterial scaffold manufactured from silk fibroin supports the maturation and long-term survival of a functional hiPSC-RPE monolayer. This has significant implications for both in vitro disease modeling and in vivo cell replacement therapy.
Collapse
Affiliation(s)
- Chad A Galloway
- Department of Ophthalmology (Flaum Eye Institute), University of Rochester, Rochester, New York, United States.,Department of Biomedical Genetics, University of Rochester, Rochester, New York, United States
| | - Sonal Dalvi
- Department of Ophthalmology (Flaum Eye Institute), University of Rochester, Rochester, New York, United States.,Department of Biomedical Genetics, University of Rochester, Rochester, New York, United States
| | - Audra M A Shadforth
- Queensland Eye Institute, South Brisbane, Queensland, Australia.,School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia.,Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Shuko Suzuki
- Queensland Eye Institute, South Brisbane, Queensland, Australia
| | - Molly Wilson
- Waisman Center, University of Wisconsin, Madison, Wisconsin, United States
| | - David Kuai
- Waisman Center, University of Wisconsin, Madison, Wisconsin, United States
| | - Ali Hashim
- Department of Ophthalmology (Flaum Eye Institute), University of Rochester, Rochester, New York, United States.,Department of Biomedical Genetics, University of Rochester, Rochester, New York, United States
| | - Leslie A MacDonald
- Department of Ophthalmology (Flaum Eye Institute), University of Rochester, Rochester, New York, United States.,Department of Biomedical Genetics, University of Rochester, Rochester, New York, United States
| | - David M Gamm
- Waisman Center, University of Wisconsin, Madison, Wisconsin, United States.,Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, Wisconsin, United States.,McPherson Eye Research Institute, University of Wisconsin, Madison, Wisconsin, United States
| | - Damien G Harkin
- Queensland Eye Institute, South Brisbane, Queensland, Australia.,School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia.,Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Ruchira Singh
- Department of Ophthalmology (Flaum Eye Institute), University of Rochester, Rochester, New York, United States.,Department of Biomedical Genetics, University of Rochester, Rochester, New York, United States.,Center for Visual Science, University of Rochester, Rochester, New York, United States.,Univeristy of Rochester Stem Cell and Regenerative Medicine Institute, Rochester, New York, United States
| |
Collapse
|
23
|
Harkin DG, Dunphy SE, Shadforth AMA, Dawson RA, Walshe J, Zakaria N. Mounting of Biomaterials for Use in Ophthalmic Cell Therapies. Cell Transplant 2018; 26:1717-1732. [PMID: 29338382 PMCID: PMC5784520 DOI: 10.1177/0963689717723638] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
When used as scaffolds for cell therapies, biomaterials often present basic handling and logistical problems for scientists and surgeons alike. The quest for an appropriate mounting device for biomaterials is therefore a significant and common problem. In this review, we provide a detailed overview of the factors to consider when choosing an appropriate mounting device including those experienced during cell culture, quality assurance, and surgery. By way of example, we draw upon our combined experience in developing epithelial cell therapies for the treatment of eye diseases. We discuss commercially available options for achieving required goals and provide a detailed analysis of 4 experimental designs developed within our respective laboratories in Australia, the United Kingdom, and Belgium.
Collapse
Affiliation(s)
- Damien G Harkin
- 1 School of Biomedical Sciences and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia.,2 Queensland Eye Institute, South Brisbane, Queensland, Australia
| | - Siobhan E Dunphy
- 3 Division of Clinical Neuroscience, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom.,4 Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College, Dublin, Ireland
| | - Audra M A Shadforth
- 1 School of Biomedical Sciences and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia.,2 Queensland Eye Institute, South Brisbane, Queensland, Australia
| | - Rebecca A Dawson
- 1 School of Biomedical Sciences and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia.,2 Queensland Eye Institute, South Brisbane, Queensland, Australia
| | - Jennifer Walshe
- 2 Queensland Eye Institute, South Brisbane, Queensland, Australia
| | - Nadia Zakaria
- 5 Division of Ophthalmology, Center for Cell Therapy and Regenerative Medicine, University Hospital Antwerp, Edegem, Belgium.,6 Department of Ophthalmology, Visual Optics and Visual Rehabilitation, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
24
|
Balagholi S, Rezaei Kanavi M, Alizadeh S, Dabbaghi R, Karami S, Kheiri B, Daftarian N. Effects of fibrin glue as a three-dimensional scaffold in cultivated adult human retinal pigment epithelial cells. J Biomater Appl 2018; 33:514-526. [DOI: 10.1177/0885328218799506] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This study was conducted to examine morphological, genotypic, and phenotypic alterations occurring in cultured adult human retinal pigment epithelial cells when encapsulated with different concentrations of fibrin glue. Cultivated adult human retinal pigment epithelial cells were encapsulated with different concentrations of fibrin glue, namely FG1 (42 mg/dl), FG2 (84 mg/dl), FG3 (124 mg/dl), FG4 (210 mg/dl), followed by the evaluation of genetic and cytomorphological changes and protein expression. Cultured adult human retinal pigment epithelial cells showed dendritiform morphology during the early days of encapsulation with fibrin glue. Moreover, an increasing inhibitory effect on cell growth was observed with increasing concentrations of fibrin glue. At the transcriptional level, the expression of MMP2, PAX6, and ITGB1 in FG1-encapsulated cells was significantly higher than that in other treated groups; however, the expression of ACTA2 was lower in all fibrin glue-encapsulated groups compared to that in the controls. Immunocytochemistry showed that FG2-encapsulated cells expressed cytokeratin 8/18, RPE65, and ZO-1 proteins, but not PAX6. In conclusion, fibrin glue at a concentration of 84 mg/dl allows proper encapsulation of adult human retinal pigment epithelial cells, while preserving the morphometric, genotypic, and phenotypic features of the cells. This three-dimensional biopolymer can be considered a reliable vehicle for retinal pigment epithelium cell transplantation in cell-based therapies.
Collapse
Affiliation(s)
- Sahar Balagholi
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mozhgan Rezaei Kanavi
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shaban Alizadeh
- Department of Hematology, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Rasul Dabbaghi
- Department of Hematology, Faculty of Medical sciences, Tarbiat Modares University, Tehran, Iran
| | - Samira Karami
- Department of Hematology, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Kheiri
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Narsis Daftarian
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Surrao DC, Greferath U, Chau YQ, Skabo SJ, Huynh M, Shelat KJ, Limnios IJ, Fletcher EL, Liu Q. Design, development and characterization of synthetic Bruch's membranes. Acta Biomater 2017; 64:357-376. [PMID: 28951331 DOI: 10.1016/j.actbio.2017.09.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 09/13/2017] [Accepted: 09/22/2017] [Indexed: 12/30/2022]
Abstract
Age-related macular degeneration (AMD) is a leading cause of blindness, and dry AMD has no effective treatment. Retinal constructs comprising retinal pigment epithelium (RPE) cells supported by electrospun scaffolds have been investigated to treat dry AMD. However, electrospun scaffolds studied to-date do not mimic the structural microenvironment of human Bruch's membrane (BM), essential for native-like RPE monolayers. The aim of this study was to develop a structurally biomimetic scaffold designed to support a functional RPE monolayer, comprising porous, electrospun nanofibrous membranes (ENMs), coated with laminin, mimicking the inner collagenous layer (ICL) and basal RPE lamina respectively, the cell supporting layers of the BM. In vitro evaluation showed 70nm PLLA ENMs adsorbed high amounts of laminin and supported functional RPE monolayers, exhibiting 3D polygonal-cobblestone morphology, apical microvilli, basal infoldings, high transepithelial resistance (TER), phagocytic activity and expression of signature RPE markers. 70nm PLLA ENMs were successfully implanted into the subretinal space of RCS-rdy+p+/LAV rats, also commonly know as rdy rats. At week 4, in the absence of immunosuppressants, implanted PLLA ENMs were surrounded by a significantly low number of activated microglial cells, compared to week 1, indicating no adverse long-term immune response. In conclusion, we successfully designed and tested ENMs emulating the RPE cell supporting layers of the BM, and found 70nm PLLA ENMs to be best suited as scaffolds for fabricating retinal constructs. STATEMENT OF SIGNIFICANCE Age related macular degeneration (AMD) is a leading cause of vision loss in the developed world, with an increasing number of people suffering from blindness or severe visual impairment. Transplantation of retinal pigment epithelium (RPE) cells supported on a synthetic, biomimetic-like Bruch's membrane (BM) is considered a promising treatment. However, the synthetic scaffolds used do not mimic the microenvironment of the RPE cell supporting layers, required for the development of a functional RPE monolayer. This study indicated that porous, laminin coated, 70nm PLLA ENMs supported functional RPE monolayers, exhibiting 3D polygonal-cobblestone morphology, apical microvilli, basal infoldings, high transepithelial resistance (TER), phagocytic activity and expression of signature RPE markers. These findings indicate the potential clinical use of porous, laminin coated, 70nm PLLA ENMs in fabricating retinal constructs aimed at treating dry AMD.
Collapse
Affiliation(s)
- Denver C Surrao
- Clem Jones Research Centre for Regenerative Medicine, Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD 4229, Australia.
| | - Ursula Greferath
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Yu-Qian Chau
- Clem Jones Research Centre for Regenerative Medicine, Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD 4229, Australia
| | - Stuart J Skabo
- Clem Jones Research Centre for Regenerative Medicine, Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD 4229, Australia
| | - Mario Huynh
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Kinnari J Shelat
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia; Australian National Fabrication Facility (ANFF), Queensland Node, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Ioannis J Limnios
- Clem Jones Research Centre for Regenerative Medicine, Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD 4229, Australia
| | - Erica L Fletcher
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Qin Liu
- Clem Jones Research Centre for Regenerative Medicine, Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD 4229, Australia
| |
Collapse
|
26
|
Abstract
Purpose of review Progress in stem cell research for blinding diseases over the past decade is now being applied to patients with retinal degenerative diseases and soon perhaps, glaucoma. However, the field still has much to learn about the conversion of stem cells into various retinal cell types, and the potential delivery methods that will be required to optimize the clinical efficacy of stem cells delivered into the eye. Recent findings Recent groundbreaking human clinical trials have demonstrated both the opportunities and current limitations of stem cell transplantation for retinal diseases. New progress in developing in vitro retinal organoids, coupled with the maturation of bio-printing technology, and non-invasive high-resolution imaging have created new possibilities for repairing and regenerating the diseased retina and rigorously validating its clinical impact in vivo. Summary While promising progress is being made, meticulous clinical trials with cells derived using good manufacturing practice, novel surgical methods, and improved methods to derive all of the neuronal cell types present in the retina will be indispensable for developing stem cell transplantation as a paradigm shift for the treatment of blinding diseases.
Collapse
|
27
|
Miyagishima KJ, Wan Q, Miller SS, Bharti K. A basis for comparison: sensitive authentication of stem cell derived RPE using physiological responses of intact RPE monolayers. STEM CELL AND TRANSLATIONAL INVESTIGATION 2017; 4:e1497. [PMID: 28286868 PMCID: PMC5341611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The retinal pigment epithelium (RPE) is a monolayer of highly specialized cells that help maintain the chemical composition of its surrounding subretinal and choroidal extracellular spaces. Retinal cells (photoreceptors in particular), RPE, and choroidal endothelial cells together help ensure a homeostatically stable metabolic environment with exquisitely sensitive functional responses to light. Aging and disease of the RPE impairs its supportive functions contributing to the progressive loss of photoreceptors and vision. The prevalence of RPE associated retinal degenerations has prompted researchers to develop new therapies aimed at replacing the affected RPE with induced pluripotent stem cell (iPSC) or embryonic stem cell (ESC) derived RPE. Despite recent attempts to characterize stem cell derived RPE and to truly authenticate RPE for clinical applications, there remains a significant unmet need to explore the heterogeneity resulting from donor to donor variation as well as the variations inherent in the current processes of cell manufacture. Additionally, it remains unknown whether the starting cell type influences the resulting RPE phenotype following reprogramming and differentiation. To address these questions, we performed a comprehensive evaluation (genomic, structural, and functional) of 15 iPSC derived RPE originating from different donors and tissues and compiled a reference data set for the authentication of iPSC-derived RPE and RPE derived from other stem cell sources.
Collapse
Affiliation(s)
- Kiyoharu J. Miyagishima
- Section on Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Qin Wan
- Section on Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sheldon S. Miller
- Section on Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kapil Bharti
- Unit on Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
28
|
Al-Nawaiseh S, Thieltges F, Liu Z, Strack C, Brinken R, Braun N, Wolschendorf M, Maminishkis A, Eter N, Stanzel BV. A Step by Step Protocol for Subretinal Surgery in Rabbits. J Vis Exp 2016. [PMID: 27684952 PMCID: PMC5092014 DOI: 10.3791/53927] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Age related macular degeneration (AMD), retinitis pigmentosa, and other RPE related diseases are the most common causes for irreversible loss of vision in adults in industrially developed countries. RPE transplantation appears to be a promising therapy, as it may replace dysfunctional RPE, restore its function, and thereby vision. Here we describe a method for transplanting a cultured RPE monolayer on a scaffold into the subretinal space (SRS) of rabbits. After vitrectomy xenotransplants were delivered into the SRS using a custom made shooter consisting of a 20-gauge metallic nozzle with a polytetrafluoroethylene (PTFE) coated plunger. The current technique evolved in over 150 rabbit surgeries over 6 years. Post-operative follow-up can be obtained using non-invasive and repetitive in vivo imaging such as spectral domain optical coherence tomography (SD-OCT) followed by perfusion-fixed histology. The method has well-defined steps for easy learning and high success rate. Rabbits are considered a large eye animal model useful in preclinical studies for clinical translation. In this context rabbits are a cost-efficient and perhaps convenient alternative to other large eye animal models.
Collapse
Affiliation(s)
| | | | - Zengping Liu
- Department of Ophthalmology, University of Bonn; Department of Ophthalmology, National University of Singapore
| | | | | | | | | | - Arvydas Maminishkis
- Section on Epithelial and Retinal Physiology and Disease, National Eye Institute/National Institutes of Health
| | - Nicole Eter
- Department of Ophthalmology, University of Münster
| | - Boris V Stanzel
- Department of Ophthalmology, University of Bonn; Surgical Retina Department, Singapore National Eye Centre;
| |
Collapse
|
29
|
Hotaling NA, Khristov V, Wan Q, Sharma R, Jha BS, Lotfi M, Maminishkis A, Simon CG, Bharti K. Nanofiber Scaffold-Based Tissue-Engineered Retinal Pigment Epithelium to Treat Degenerative Eye Diseases. J Ocul Pharmacol Ther 2016; 32:272-85. [PMID: 27110730 PMCID: PMC4904235 DOI: 10.1089/jop.2015.0157] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/24/2016] [Indexed: 12/16/2022] Open
Abstract
Clinical-grade manufacturing of a functional retinal pigment epithelium (RPE) monolayer requires reproducing, as closely as possible, the natural environment in which RPE grows. In vitro, this can be achieved by a tissue engineering approach, in which the RPE is grown on a nanofibrous biological or synthetic scaffold. Recent research has shown that nanofiber scaffolds perform better for cell growth and transplantability compared with their membrane counterparts and that the success of the scaffold in promoting cell growth/function is not heavily material dependent. With these strides, the field has advanced enough to begin to consider implementation of one, or a combination, of the tissue engineering strategies discussed herein. In this study, we review the current state of tissue engineering research for in vitro culture of RPE/scaffolds and the parameters for optimal scaffold design that have been uncovered during this research. Next, we discuss production methods and manufacturers that are capable of producing the nanofiber scaffolds in such a way that would be biologically, regulatory, clinically, and commercially viable. Then, a discussion of how the scaffolds could be characterized, both morphologically and mechanically, to develop a testing process that is viable for regulatory screening is performed. Finally, an example of a tissue-engineered RPE/scaffold construct is given to provide the reader a framework for understanding how these pieces could fit together to develop a tissue-engineered RPE/scaffold construct that could pass regulatory scrutiny and can be commercially successful.
Collapse
Affiliation(s)
- Nathan A. Hotaling
- Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, Maryland
- Unit on Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Vladimir Khristov
- Section of Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Qin Wan
- Section of Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Ruchi Sharma
- Unit on Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Balendu Shekhar Jha
- Unit on Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Mostafa Lotfi
- Section of Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Arvydas Maminishkis
- Section of Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Carl G. Simon
- Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, Maryland
| | - Kapil Bharti
- Unit on Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
30
|
Song MJ, Bharti K. Looking into the future: Using induced pluripotent stem cells to build two and three dimensional ocular tissue for cell therapy and disease modeling. Brain Res 2016; 1638:2-14. [PMID: 26706569 PMCID: PMC4837038 DOI: 10.1016/j.brainres.2015.12.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 11/24/2015] [Accepted: 12/08/2015] [Indexed: 01/02/2023]
Abstract
Retinal degenerative diseases are the leading cause of irreversible vision loss in developed countries. In many cases the diseases originate in the homeostatic unit in the back of the eye that contains the retina, retinal pigment epithelium (RPE) and the choriocapillaris. RPE is a central and a critical component of this homeostatic unit, maintaining photoreceptor function and survival on the apical side and choriocapillaris health on the basal side. In diseases like age-related macular degeneration (AMD), it is thought that RPE dysfunctions cause disease-initiating events and as the RPE degenerates photoreceptors begin to die and patients start loosing vision. Patient-specific induced pluripotent stem (iPS) cell-derived RPE provides direct access to a patient's genetics and allow the possibility of identifying the initiating events of RPE-associated degenerative diseases. Furthermore, iPS cell-derived RPE cells are being tested as a potential cell replacement in disease stages with RPE atrophy. In this article we summarize the recent progress in the field of iPS cell-derived RPE "disease modeling" and cell therapies and also discuss the possibilities of developing a model of the entire homeostatic unit to aid in studying disease processes in the future. This article is part of a Special Issue entitled SI: PSC and the brain.
Collapse
Affiliation(s)
- Min Jae Song
- Unit on Ocular and Stem Cell Translational Research National Eye Institute, 10 Center Drive, Room 10B10, Bethesda, MD 20892, United States
| | - Kapil Bharti
- Unit on Ocular and Stem Cell Translational Research National Eye Institute, 10 Center Drive, Room 10B10, Bethesda, MD 20892, United States.
| |
Collapse
|
31
|
Chamling X, Sluch VM, Zack DJ. The Potential of Human Stem Cells for the Study and Treatment of Glaucoma. Invest Ophthalmol Vis Sci 2016; 57:ORSFi1-6. [PMID: 27116666 PMCID: PMC5110236 DOI: 10.1167/iovs.15-18590] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 01/05/2016] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Currently, the only available and approved treatments for glaucoma are various pharmacologic, laser-based, and surgical procedures that lower IOP. Although these treatments can be effective, they are not always sufficient, and they cannot restore vision that has already been lost. The goal of this review is to briefly assess current developments in the application of stem cell biology to the study and treatment of glaucoma and other forms of optic neuropathy. METHODS A combined literature review and summary of the glaucoma-related discussion at the 2015 "Sight Restoration Through Stem Cell Therapy" meeting that was sponsored by the Ocular Research Symposia Foundation (ORSF). RESULTS Ongoing advancements in basic and eye-related developmental biology have enabled researchers to direct murine and human stem cells along specific developmental paths and to differentiate them into a variety of ocular cell types of interest. The most advanced of these efforts involve the differentiation of stem cells into retinal pigment epithelial cells, work that has led to the initiation of several human trials. More related to the glaucoma field, there have been recent advances in developing protocols for differentiation of stem cells into trabecular meshwork and retinal ganglion cells. Additionally, efforts are being made to generate stem cell-derived cells that can be used to secrete neuroprotective factors. CONCLUSIONS Advancing stem cell technology provides opportunities to improve our understanding of glaucoma-related biology and develop models for drug development, and offers the possibility of cell-based therapies to restore sight to patients who have already lost vision.
Collapse
Affiliation(s)
- Xitiz Chamling
- Department of Ophthalmology Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Valentin M. Sluch
- Department of Ophthalmology Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Donald J. Zack
- Department of Ophthalmology Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
32
|
Di Foggia V, Makwana P, Ali RR, Sowden JC. Induced Pluripotent Stem Cell Therapies for Degenerative Disease of the Outer Retina: Disease Modeling and Cell Replacement. J Ocul Pharmacol Ther 2016; 32:240-52. [PMID: 27027805 DOI: 10.1089/jop.2015.0143] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Stem cell therapies are being explored as potential treatments for retinal disease. How to replace neurons in a degenerated retina presents a continued challenge for the regenerative medicine field that, if achieved, could restore sight. The major issues are: (i) the source and availability of donor cells for transplantation; (ii) the differentiation of stem cells into the required retinal cells; and (iii) the delivery, integration, functionality, and survival of new cells in the host neural network. This review considers the use of induced pluripotent stem cells (iPSC), currently under intense investigation, as a platform for cell transplantation therapy. Moreover, patient-specific iPSC are being developed for autologous cell transplantation and as a tool for modeling specific retinal diseases, testing gene therapies, and drug screening.
Collapse
Affiliation(s)
- Valentina Di Foggia
- 1 UCL Institute of Child Health, University College London , London, United Kingdom
| | - Priyanka Makwana
- 1 UCL Institute of Child Health, University College London , London, United Kingdom
| | - Robin R Ali
- 2 UCL Institute of Ophthalmology , London, United Kingdom
| | - Jane C Sowden
- 1 UCL Institute of Child Health, University College London , London, United Kingdom
| |
Collapse
|
33
|
Ouyang H, Goldberg JL, Chen S, Li W, Xu GT, Li W, Zhang K, Nussenblatt RB, Liu Y, Xie T, Chan CC, Zack DJ. Ocular Stem Cell Research from Basic Science to Clinical Application: A Report from Zhongshan Ophthalmic Center Ocular Stem Cell Symposium. Int J Mol Sci 2016; 17:415. [PMID: 27102165 PMCID: PMC4813266 DOI: 10.3390/ijms17030415] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 03/17/2016] [Accepted: 03/17/2016] [Indexed: 12/16/2022] Open
Abstract
Stem cells hold promise for treating a wide variety of diseases, including degenerative disorders of the eye. The eye is an ideal organ for stem cell therapy because of its relative immunological privilege, surgical accessibility, and its being a self-contained system. The eye also has many potential target diseases amenable to stem cell-based treatment, such as corneal limbal stem cell deficiency, glaucoma, age-related macular degeneration (AMD), and retinitis pigmentosa (RP). Among them, AMD and glaucoma are the two most common diseases, affecting over 200 million people worldwide. Recent results on the clinical trial of retinal pigment epithelial (RPE) cells from human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) in treating dry AMD and Stargardt’s disease in the US, Japan, England, and China have generated great excitement and hope. This marks the beginning of the ocular stem cell therapy era. The recent Zhongshan Ophthalmic Center Ocular Stem Cell Symposium discussed the potential applications of various stem cell types in stem cell-based therapies, drug discoveries and tissue engineering for treating ocular diseases.
Collapse
Affiliation(s)
- Hong Ouyang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China.
| | - Jeffrey L Goldberg
- Department of Ophthalmology, Stanford University, Palo Alto, CA 94303, USA.
| | - Shuyi Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China.
| | - Wei Li
- Unit on Retinal Neurophysiology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Guo-Tong Xu
- Department of Ophthalmology, Tongji University, Shanghai 200092, China.
| | - Wei Li
- Department of Ophthalmology, Xiamen University, Xiamen 361005, China.
| | - Kang Zhang
- Department of Ophthalmology, University of California San Diego, San Diego, CA 92093, USA.
| | - Robert B Nussenblatt
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Yizhi Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China.
| | - Ting Xie
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA.
| | - Chi-Chao Chan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China.
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Donald J Zack
- Wilmer Ophthalmological Institute, Johns Hopkins University, Baltimore, MD 21231, USA.
| |
Collapse
|
34
|
Abstract
Induced pluripotent stem (iPS) cells have great potential in regenerative medicine, including cell replacement therapies and disease modelling in vitro. However, with this potential comes several challenges, including clinical safety, reprogramming and differentiation efficiency, and compromised functionality of differentiated cell types after transplantation. Many of these issues arise from imprecise control of cell fate. With large-scale sequencing and genome-editing technologies we can now precisely manipulate the genome, which has expanded our knowledge of functional cell types and cell identity. These technologies may improve our efforts in generating iPS-derived therapeutic cells and in development of therapies for human diseases.
Collapse
|
35
|
Shadforth AMA, Suzuki S, Alzonne R, Edwards GA, Richardson NA, Chirila TV, Harkin DG. Incorporation of Human Recombinant Tropoelastin into Silk Fibroin Membranes with the View to Repairing Bruch's Membrane. J Funct Biomater 2015; 6:946-62. [PMID: 26389960 PMCID: PMC4598686 DOI: 10.3390/jfb6030946] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 08/18/2015] [Accepted: 09/11/2015] [Indexed: 11/24/2022] Open
Abstract
Bombyx mori silk fibroin membranes provide a potential delivery vehicle for both cells and extracellular matrix (ECM) components into diseased or injured tissues. We have previously demonstrated the feasibility of growing retinal pigment epithelial cells (RPE) on fibroin membranes with the view to repairing the retina of patients afflicted with age-related macular degeneration (AMD). The goal of the present study was to investigate the feasibility of incorporating the ECM component elastin, in the form of human recombinant tropoelastin, into these same membranes. Two basic strategies were explored: (1) membranes prepared from blended solutions of fibroin and tropoelastin; and (2) layered constructs prepared from sequentially cast solutions of fibroin, tropoelastin, and fibroin. Optimal conditions for RPE attachment were achieved using a tropoelastin-fibroin blend ratio of 10 to 90 parts by weight. Retention of tropoelastin within the blend and layered constructs was confirmed by immunolabelling and Fourier-transform infrared spectroscopy (FTIR). In the layered constructs, the bulk of tropoelastin was apparently absorbed into the initially cast fibroin layer. Blend membranes displayed higher elastic modulus, percentage elongation, and tensile strength (p < 0.01) when compared to the layered constructs. RPE cell response to fibroin membranes was not affected by the presence of tropoelastin. These findings support the potential use of fibroin membranes for the co-delivery of RPE cells and tropoelastin.
Collapse
Affiliation(s)
- Audra M A Shadforth
- Queensland Eye Institute, 140 Melbourne Street, South Brisbane, Queensland 4101, Australia.
- School of Biomedical Sciences and Institute of Health & Biomedical Innovation, Queensland University of Technology, 2 George Street, Brisbane, Queensland 4001, Australia.
| | - Shuko Suzuki
- Queensland Eye Institute, 140 Melbourne Street, South Brisbane, Queensland 4101, Australia.
| | - Raphaelle Alzonne
- Queensland Eye Institute, 140 Melbourne Street, South Brisbane, Queensland 4101, Australia.
| | - Grant A Edwards
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia.
| | - Neil A Richardson
- Queensland Eye Institute, 140 Melbourne Street, South Brisbane, Queensland 4101, Australia.
- School of Biomedical Sciences and Institute of Health & Biomedical Innovation, Queensland University of Technology, 2 George Street, Brisbane, Queensland 4001, Australia.
| | - Traian V Chirila
- Queensland Eye Institute, 140 Melbourne Street, South Brisbane, Queensland 4101, Australia.
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia.
- Science and Engineering Faculty, Queensland University of Technology, Brisbane, Queensland 4001, Australia.
- Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, Queensland 4029, Australia.
- Faculty of Science, University of Western Australia, Crawley, Western Australia 6009, Australia.
| | - Damien G Harkin
- Queensland Eye Institute, 140 Melbourne Street, South Brisbane, Queensland 4101, Australia.
- School of Biomedical Sciences and Institute of Health & Biomedical Innovation, Queensland University of Technology, 2 George Street, Brisbane, Queensland 4001, Australia.
| |
Collapse
|