1
|
MacLaren RE, Audo I, Fischer MD, Huckfeldt RM, Lam BL, Pennesi ME, Sisk R, Gow JA, Li J, Zhu K, Tsang SF. An Open-Label Phase II Study Assessing the Safety of Bilateral, Sequential Administration of Retinal Gene Therapy in Participants with Choroideremia: The GEMINI Study. Hum Gene Ther 2024; 35:564-575. [PMID: 38970425 DOI: 10.1089/hum.2024.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2024] Open
Abstract
Choroideremia, an incurable, progressive retinal degeneration primarily affecting young men, leads to sight loss. GEMINI was a multicenter, open-label, prospective, two-period, interventional Phase II study assessing the safety of bilateral sequential administration of timrepigene emparvovec, a gene therapy, in adult males with genetically confirmed choroideremia (NCT03507686, ClinicalTrials.gov). Timrepigene emparvovec is an adeno-associated virus serotype 2 vector encoding the cDNA of Rab escort protein 1, augmented by a downstream woodchuck hepatitis virus post-transcriptional regulatory element. Up to 0.1 mL of timrepigene emparvovec, containing 1 × 1011 vector genomes, was administered by subretinal injection following vitrectomy and retinal detachment. The second eye was treated after an intrasurgery window of <6, 6-12, or >12 months. Each eye was followed at up to nine visits over 12 months. Overall, 66 participants received timrepigene emparvovec, and 53 completed the study. Visual acuity (VA) was generally maintained in both eyes, independent of intrasurgery window duration, even after bilateral retinal detachment and subretinal injection. Bilateral treatment was well tolerated, with predominantly mild or moderate treatment-emergent adverse events (TEAEs) and a low rate of serious surgical complications (7.6%). Retinal inflammation TEAEs were reported in 45.5% of participants, with similar rates in both eyes; post hoc analyses found that these were not associated with clinically significant vision loss at month 12 versus baseline. Two participants (3.0%) reported serious noninfective retinitis. Prior timrepigene emparvovec exposure did not increase the risk of serious TEAEs or serious ocular TEAEs upon injection of the second eye; furthermore, no systemic immune reaction or inoculation effect was observed. Presence of antivector neutralizing antibodies at baseline was potentially associated with a higher percentage of TEAEs related to ocular inflammation or reduced VA after injection of the first eye. The GEMINI study results may inform decisions regarding bilateral sequential administration of other gene therapies for retinal diseases.
Collapse
Affiliation(s)
- Robert E MacLaren
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- Oxford NIHR Biomedical Research Centre, Oxford, United Kingdom
| | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and INSERM-DGOS CIC 1423, Paris, France
| | - M Dominik Fischer
- University Eye Hospital, Centre for Ophthalmology, University Hospital Tübingen, Tübingen, Germany
| | - Rachel M Huckfeldt
- MEE Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| | - Byron L Lam
- Bascom Palmer Eye Institute, University of Miami, Miami, Florida, USA
| | - Mark E Pennesi
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Robert Sisk
- Cincinnati Eye Institute, Blue Ash, Ohio, USA
| | | | - Jiang Li
- Biogen Inc., Cambridge, Massachusetts, USA
| | - Kan Zhu
- Biogen Inc., Cambridge, Massachusetts, USA
| | | |
Collapse
|
2
|
Hensman J, Hahn LC, van Schooneveld MJ, Diederen RMH, Ten Brink JB, Florijn RJ, Bergen AA, Strubbe I, Heutinck P, van Genderen MM, van den Born LI, Thiadens AA, de Zaeytijd J, Leroy BP, Hoyng CB, Boon CJF. Efficacy of Carbonic Anhydrase Inhibitors on Cystoid Fluid Collections and Visual Acuity in Patients with X-Linked Retinoschisis. Ophthalmol Retina 2024; 8:600-606. [PMID: 38104928 DOI: 10.1016/j.oret.2023.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/22/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
PURPOSE To date, there is no standard treatment regimen for carbonic anhydrase inhibitors (CAIs) in X-linked retinoschisis (XLRS) patients. This retrospective study aims to evaluate the efficacy of CAIs on visual acuity and cystoid fluid collections (CFC) in XRLS patients in Dutch and Belgian tertiary referral centers. DESIGN Retrospective cohort study. PARTICIPANTS Forty-two patients with XLRS. METHODS In total, 42 patients were enrolled. To be included, patients had to have previous treatment with an oral CAI (acetazolamide), a topical CAI (brinzolamide/dorzolamide), or a combination of an oral and a topical CAI for at least 4 consecutive weeks. We evaluated the effect of the CAI on best-corrected visual acuity (BCVA) and central foveal thickness (CFT) on OCT. MAIN OUTCOME MEASURES Central foveal thickness and BCVA. RESULTS The median age at the baseline visit of the patients in this cohort study was 14.7 (range, 43.6) years, with a median (interquartile range [IQR]) follow-up period of 4.0 (2.2-5.2) years. During the follow-up period, 25 patients were treated once with an oral CAI (60%), 24 patients were treated once with a topical CAI (57%), and 11 patients were treated once with a combination of both topical and oral CAI (26%). We observed a significant reduction of CFT for oral CAI by 14.37 μm per 100 mg per day (P < 0.001; 95% confidence interval [CI], -19.62 to -9.10 μm) and for topical CAI by 7.52 μm per drop per day (P = 0.017; 95% CI, -13.67 to -1.32 μm). The visual acuity changed significantly while on treatment with oral CAI by -0.0059 logMAR per 100 mg (P = 0.008; 95% CI, -0.010 to -0.0013 logMAR). Seven patients (17%) had side effects leading to treatment discontinuation. CONCLUSIONS Our data indicate that treatment with (oral) CAI may be beneficial for short-term management of CFC in patients with XLRS. Despite a significant reduction in CFT, the change in visual acuity was modest and not of clinical significance. Nonetheless, the anatomic improvement of the central retina in these patients may be of value to create an optimal retinal condition for future potential treatment options such as gene therapy. FINANCIAL DISCLOSURE(S) The authors have no proprietary or commercial interest in any materials discussed in this article.
Collapse
Affiliation(s)
- Jonathan Hensman
- Department of Ophthalmology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Leo C Hahn
- Department of Ophthalmology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Mary J van Schooneveld
- Department of Ophthalmology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Roselie M H Diederen
- Department of Ophthalmology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Jacoline B Ten Brink
- Department of Human Genetics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Ralph J Florijn
- Department of Human Genetics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Arthur A Bergen
- Department of Human Genetics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Ine Strubbe
- Department of Ophthalmology, Ghent University Hospital, Ghent, Belgium
| | - Pam Heutinck
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Maria M van Genderen
- Bartiméus, Diagnostic Centre for Complex Visual Disorders, Zeist, The Netherlands
| | | | - Alberta A Thiadens
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Julie de Zaeytijd
- Department of Ophthalmology, Ghent University Hospital, Ghent, Belgium
| | - Bart P Leroy
- Department of Ophthalmology, Ghent University Hospital, Ghent, Belgium
| | - Carel B Hoyng
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Camiel J F Boon
- Department of Ophthalmology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands; Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
3
|
Brar AS, Parameswarappa DC, Takkar B, Narayanan R, Jalali S, Mandal S, Fujinami K, Padhy SK. Gene Therapy for Inherited Retinal Diseases: From Laboratory Bench to Patient Bedside and Beyond. Ophthalmol Ther 2024; 13:21-50. [PMID: 38113023 PMCID: PMC10776519 DOI: 10.1007/s40123-023-00862-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 11/23/2023] [Indexed: 12/21/2023] Open
Abstract
This comprehensive review provides a thorough examination of inherited retinal diseases (IRDs), encompassing their classification, genetic underpinnings, and the promising landscape of gene therapy trials. IRDs, a diverse group of genetic conditions causing vision loss through photoreceptor cell death, are explored through various angles, including inheritance patterns, gene involvement, and associated systemic disorders. The focal point is gene therapy, which offers hope for halting or even reversing the progression of IRDs. The review highlights ongoing clinical trials spanning retinal cell replacement, neuroprotection, pharmacological interventions, and optogenetics. While these therapies hold tremendous potential, they face challenges like timing optimization, standardized assessment criteria, inflammation management, vector refinement, and raising awareness among vision scientists. Additionally, translating gene therapy success into widespread adoption and addressing cost-effectiveness are crucial challenges to address. Continued research and clinical trials are essential to fully harness gene therapy's potential in treating IRDs and enhancing the lives of affected individuals.
Collapse
Affiliation(s)
- Anand Singh Brar
- Anant Bajaj Retina Institute, LV Prasad Eye Institute, Mithu Tulsi Chanrai Campus, Bhubaneswar, 751024, India
| | - Deepika C Parameswarappa
- Anant Bajaj Retina Institute, LV Prasad Eye Institute, Kallam Anji Reddy Campus, Hyderabad, 500034, India
| | - Brijesh Takkar
- Anant Bajaj Retina Institute, LV Prasad Eye Institute, Kallam Anji Reddy Campus, Hyderabad, 500034, India
| | - Raja Narayanan
- Anant Bajaj Retina Institute, LV Prasad Eye Institute, Kallam Anji Reddy Campus, Hyderabad, 500034, India
| | - Subhadra Jalali
- Anant Bajaj Retina Institute, LV Prasad Eye Institute, Kallam Anji Reddy Campus, Hyderabad, 500034, India
| | - Sohini Mandal
- Dr Rajendra Prasad Center for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Kaoru Fujinami
- Laboratory of Visual Physiology, Division of Vision Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, 152-8902, Japan
| | - Srikanta Kumar Padhy
- Anant Bajaj Retina Institute, LV Prasad Eye Institute, Mithu Tulsi Chanrai Campus, Bhubaneswar, 751024, India.
| |
Collapse
|
4
|
Wang T, Li H, Pu T, Yang L. Microsurgery Robots: Applications, Design, and Development. SENSORS (BASEL, SWITZERLAND) 2023; 23:8503. [PMID: 37896597 PMCID: PMC10611418 DOI: 10.3390/s23208503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 10/07/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023]
Abstract
Microsurgical techniques have been widely utilized in various surgical specialties, such as ophthalmology, neurosurgery, and otolaryngology, which require intricate and precise surgical tool manipulation on a small scale. In microsurgery, operations on delicate vessels or tissues require high standards in surgeons' skills. This exceptionally high requirement in skills leads to a steep learning curve and lengthy training before the surgeons can perform microsurgical procedures with quality outcomes. The microsurgery robot (MSR), which can improve surgeons' operation skills through various functions, has received extensive research attention in the past three decades. There have been many review papers summarizing the research on MSR for specific surgical specialties. However, an in-depth review of the relevant technologies used in MSR systems is limited in the literature. This review details the technical challenges in microsurgery, and systematically summarizes the key technologies in MSR with a developmental perspective from the basic structural mechanism design, to the perception and human-machine interaction methods, and further to the ability in achieving a certain level of autonomy. By presenting and comparing the methods and technologies in this cutting-edge research, this paper aims to provide readers with a comprehensive understanding of the current state of MSR research and identify potential directions for future development in MSR.
Collapse
Affiliation(s)
- Tiexin Wang
- ZJU-UIUC Institute, International Campus, Zhejiang University, Haining 314400, China; (T.W.); (H.L.); (T.P.)
- School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China
| | - Haoyu Li
- ZJU-UIUC Institute, International Campus, Zhejiang University, Haining 314400, China; (T.W.); (H.L.); (T.P.)
| | - Tanhong Pu
- ZJU-UIUC Institute, International Campus, Zhejiang University, Haining 314400, China; (T.W.); (H.L.); (T.P.)
| | - Liangjing Yang
- ZJU-UIUC Institute, International Campus, Zhejiang University, Haining 314400, China; (T.W.); (H.L.); (T.P.)
- School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Mechanical Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
5
|
Luis J, Eastlake K, Lamb WDB, Limb GA, Jayaram H, Khaw PT. Cell-Based Therapies for Glaucoma. Transl Vis Sci Technol 2023; 12:23. [PMID: 37494052 PMCID: PMC10383000 DOI: 10.1167/tvst.12.7.23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 06/20/2023] [Indexed: 07/27/2023] Open
Abstract
Glaucomatous optic neuropathy (GON) is the major cause of irreversible visual loss worldwide and can result from a range of disease etiologies. The defining features of GON are retinal ganglion cell (RGC) degeneration and characteristic cupping of the optic nerve head (ONH) due to tissue remodeling, while intraocular pressure remains the only modifiable GON risk factor currently targeted by approved clinical treatment strategies. Efforts to understand the mechanisms that allow species such as the zebrafish to regenerate their retinal cells have greatly increased our understanding of regenerative signaling pathways. However, proper integration within the retina and projection to the brain by the newly regenerated neuronal cells remain major hurdles. Meanwhile, a range of methods for in vitro differentiation have been developed to derive retinal cells from a variety of cell sources, including embryonic and induced pluripotent stem cells. More recently, there has been growing interest in the implantation of glial cells as well as cell-derived products, including neurotrophins, microRNA, and extracellular vesicles, to provide functional support to vulnerable structures such as RGC axons and the ONH. These approaches offer the advantage of not relying upon the replacement of degenerated cells and potentially targeting earlier stages of disease pathogenesis. In order to translate these techniques into clinical practice, appropriate cell sourcing, robust differentiation protocols, and accurate implantation methods are crucial to the success of cell-based therapy in glaucoma. Translational Relevance: Cell-based therapies for glaucoma currently under active development include the induction of endogenous regeneration, implantation of exogenously derived retinal cells, and utilization of cell-derived products to provide functional support.
Collapse
Affiliation(s)
- Joshua Luis
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - Karen Eastlake
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - William D. B. Lamb
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - G. Astrid Limb
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - Hari Jayaram
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - Peng T. Khaw
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| |
Collapse
|
6
|
Mansouri V. X-Linked Retinitis Pigmentosa Gene Therapy: Preclinical Aspects. Ophthalmol Ther 2022; 12:7-34. [PMID: 36346573 PMCID: PMC9641696 DOI: 10.1007/s40123-022-00602-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022] Open
Abstract
The most common inherited eye disease is retinitis pigmentosa (RP). X-linked RP (XLRP) is one of the most severe types of RP, with a considerable disease burden. Patients with XLRP experience a decrease in their vision and become blind in their 4th decade of life, causing much morbidity after starting a rather normal life. Treatment of XLRP remains challenging, and current treatments are not effective enough in restoring vision. Gene therapy of XLRP, capable of restoring the functional RPGR gene, showed promising results in preclinical studies and clinical trials; however, to date, no approved product has entered the market. The development of a gene therapy product needs through preliminary assessment of the drug in animal models before administration to humans. In this article, we reviewed the genetic pathology of XLRP, along with the preclinical aspects of the XLRP gene therapy, animal models, associated assessments, and future challenges and directions.
Collapse
Affiliation(s)
- Vahid Mansouri
- Gene Therapy Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Irigoyen C, Amenabar Alonso A, Sanchez-Molina J, Rodríguez-Hidalgo M, Lara-López A, Ruiz-Ederra J. Subretinal Injection Techniques for Retinal Disease: A Review. J Clin Med 2022; 11:jcm11164717. [PMID: 36012955 PMCID: PMC9409835 DOI: 10.3390/jcm11164717] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/01/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
Inherited retinal dystrophies (IRDs) affect an estimated 1 in every 2000 people, this corresponding to nearly 2 million cases worldwide. Currently, 270 genes have been associated with IRDs, most of them altering the function of photoreceptors and retinal pigment epithelium. Gene therapy has been proposed as a potential tool for improving visual function in these patients. Clinical trials in animal models and humans have been successful in various types of IRDs. Recently, voretigene neparvovec (Luxturna®) has been approved by the US Food and Drug Administration for the treatment of biallelic mutations in the RPE65 gene. The current state of the art in gene therapy involves the delivery of various types of viral vectors into the subretinal space to effectively transduce diseased photoreceptors and retinal pigment epithelium. For this, subretinal injection is becoming increasingly popular among researchers and clinicians. To date, several approaches for subretinal injection have been described in the scientific literature, all of them effective in accessing the subretinal space. The growth and development of gene therapy give rise to the need for a standardized procedure for subretinal injection that ensures the efficacy and safety of this new approach to drug delivery. The goal of this review is to offer an insight into the current subretinal injection techniques and understand the key factors in the success of this procedure.
Collapse
Affiliation(s)
- Cristina Irigoyen
- Department of Ophthalmology, Donostia University Hospital (HUD), 20014 Donostia San-Sebastián, Spain
- Biodonostia Health Research Institute, 20014 Donostia-San Sebastián, Spain
- Department of Ophthalmology, University of the Basque Country, 48940 Leioa, Spain
| | - Asier Amenabar Alonso
- Department of Ophthalmology, Donostia University Hospital (HUD), 20014 Donostia San-Sebastián, Spain
| | - Jorge Sanchez-Molina
- Department of Ophthalmology, Donostia University Hospital (HUD), 20014 Donostia San-Sebastián, Spain
- Correspondence: ; Tel.: +34-629950276
| | | | | | | |
Collapse
|
8
|
Tichotová L, Studenovska H, Petrovski G, Popelka Š, Nemesh Y, Sedláčková M, Drutovič S, Rohiwal S, Jendelová P, Erceg S, Brymová A, Artero‐Castro A, Lytvynchuk L, Straňák Z, Ellederová Z, Motlík J, Ardan T. Advantages of nanofibrous membranes for culturing of primary RPE cells compared to commercial scaffolds. Acta Ophthalmol 2021; 100:e1172-e1185. [PMID: 34687141 DOI: 10.1111/aos.15034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 09/22/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Dysfunction of the retinal pigment epithelium (RPE) causes numerous forms of retinal degeneration. RPE replacement is a modern option to save vision. We aimed to test the results of transplanting cultured RPEs on biocompatible membranes. METHODS We cultivated porcine primary RPE cells isolated from cadaver eyes from the slaughterhouse on two types of membranes: commercial polyester scaffolds Transwell (Corning Inc., Kenneburg, ME, USA) with 0.4 µm pore size and prepared Poly (L-lactide-co-DL-lactide) (PDLLA) nanofibrous membranes with an average pore size of 0.4 µm. RESULTS Five types of assays were used for the analysis: immunocytochemistry (ICC), phagocytosis assay, Western blotting, real-time qPCR (RT-qPCR) and electron microscopy. RT-qPCR demonstrated that RPEs cultured on nanofibrous membranes have higher expressions of BEST1 (bestrophin 1), RLBP1 (retinaldehyde-binding protein 1), RPE65 (retinal pigment epithelium-specific 65 kDa protein), PAX6 (transcription factor PAX6), SOX9 (transcription factor SOX9), DCT (dopachrome tautomerase) and MITF (microphthalmia-associated transcription factor). ICC of the RPEs cultured on nanofibrous membranes showed more intensive staining of markers such as BEST1, MCT1 (monocarboxylate transporter 1), Na+ /K+ ATPase, RPE65 and acetylated tubulin in comparison with commercial ones. Additionally, the absence of α-SMA proved the stability of the RPE polarization state and the absence of epithelial-to-mesenchymal transition. RPE possessed high phagocytic activity. Electron microscopy of both membranes confirmed a confluent layer of RPE cells and their genuine morphological structure, which was comparable to native RPEs. CONCLUSIONS Retinal pigment epitheliums cultured on polylactide nanofibrous membranes improved the final quality of the cell product by having better maturation and long-term survival of the RPE monolayer compared to those cultured on commercial polyester scaffolds. PDLLA-cultured RPEs are a plausible source for the replacement of non-functioning RPEs during cell therapy.
Collapse
Affiliation(s)
- Lucie Tichotová
- Institute of Animal Physiology and Genetics Academy of Sciences of the Czech Republic Libechov Czech Republic
- Department of Cell Biology Faculty of Science Charles University Prague Czech Republic
| | - Hana Studenovska
- Institute of Macromolecular Chemistry Academy of Sciences of the Czech Republic Prague Czech Republic
| | - Goran Petrovski
- Center for Eye Research Department of Ophthalmology Oslo University Hospital and Institute for Clinical Medicine University of Oslo Oslo Norway
| | - Štěpán Popelka
- Institute of Macromolecular Chemistry Academy of Sciences of the Czech Republic Prague Czech Republic
| | - Yaroslav Nemesh
- Institute of Animal Physiology and Genetics Academy of Sciences of the Czech Republic Libechov Czech Republic
- Department of Cell Biology Faculty of Science Charles University Prague Czech Republic
| | - Miroslava Sedláčková
- Department of Histology and Embryology Faculty of Medicine Masaryk University Brno Czech Republic
| | - Saskia Drutovič
- Institute of Animal Physiology and Genetics Academy of Sciences of the Czech Republic Libechov Czech Republic
| | - Sonali Rohiwal
- Institute of Animal Physiology and Genetics Academy of Sciences of the Czech Republic Libechov Czech Republic
| | - Pavla Jendelová
- Institute of Experimental Medicine Academy of Sciences of the Czech Republic Prague Czech Republic
| | - Slaven Erceg
- Institute of Experimental Medicine Academy of Sciences of the Czech Republic Prague Czech Republic
- Stem Cell Therapies in Neurodegenerative Diseases Lab Research Center ‘Principe Felipe’ Valencia Spain
| | - Anna Brymová
- Institute of Animal Physiology and Genetics Academy of Sciences of the Czech Republic Libechov Czech Republic
- Department of Cell Biology Faculty of Science Charles University Prague Czech Republic
| | - Ana Artero‐Castro
- Stem Cell Therapies in Neurodegenerative Diseases Lab Research Center ‘Principe Felipe’ Valencia Spain
| | - Lyubomyr Lytvynchuk
- Department of Ophthalmology Justus‐Liebig‐University Giessen University Hospital Giessen and Marburg Giessen Germany
| | - Zbyněk Straňák
- Ophthalmology Department of 3rd Faculty of Medicine Charles University and University Hospital Kralovske Vinohrady Prague Czech Republic
- Third Faculty of Medicine Charles University Prague Czech Republic
| | - Zdeňka Ellederová
- Institute of Animal Physiology and Genetics Academy of Sciences of the Czech Republic Libechov Czech Republic
| | - Jan Motlík
- Institute of Animal Physiology and Genetics Academy of Sciences of the Czech Republic Libechov Czech Republic
| | - Taras Ardan
- Institute of Animal Physiology and Genetics Academy of Sciences of the Czech Republic Libechov Czech Republic
| |
Collapse
|
9
|
Chung SH, Sin TN, Ngo T, Yiu G. CRISPR Technology for Ocular Angiogenesis. Front Genome Ed 2020; 2:594984. [PMID: 34713223 PMCID: PMC8525361 DOI: 10.3389/fgeed.2020.594984] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/01/2020] [Indexed: 12/24/2022] Open
Abstract
Among genome engineering tools, Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-based approaches have been widely adopted for translational studies due to their robustness, precision, and ease of use. When delivered to diseased tissues with a viral vector such as adeno-associated virus, direct genome editing can be efficiently achieved in vivo to treat different ophthalmic conditions. While CRISPR has been actively explored as a strategy for treating inherited retinal diseases, with the first human trial recently initiated, its applications for complex, multifactorial conditions such as ocular angiogenesis has been relatively limited. Currently, neovascular retinal diseases such as retinopathy of prematurity, proliferative diabetic retinopathy, and neovascular age-related macular degeneration, which together constitute the majority of blindness in developed countries, are managed with frequent and costly injections of anti-vascular endothelial growth factor (anti-VEGF) agents that are short-lived and burdensome for patients. By contrast, CRISPR technology has the potential to suppress angiogenesis permanently, with the added benefit of targeting intracellular signals or regulatory elements, cell-specific delivery, and multiplexing to disrupt different pro-angiogenic factors simultaneously. However, the prospect of permanently suppressing physiologic pathways, the unpredictability of gene editing efficacy, and concerns for off-target effects have limited enthusiasm for these approaches. Here, we review the evolution of gene therapy and advances in adapting CRISPR platforms to suppress retinal angiogenesis. We discuss different Cas9 orthologs, delivery strategies, and different genomic targets including VEGF, VEGF receptor, and HIF-1α, as well as the advantages and disadvantages of genome editing vs. conventional gene therapies for multifactorial disease processes as compared to inherited monogenic retinal disorders. Lastly, we describe barriers that must be overcome to enable effective adoption of CRISPR-based strategies for the management of ocular angiogenesis.
Collapse
Affiliation(s)
| | | | | | - Glenn Yiu
- Department of Ophthalmology and Vision Science, University of California, Davis, Sacramento, CA, United States
| |
Collapse
|
10
|
Kahraman NS, Öner A. Umbilical cord-derived mesenchymal stem cell implantation in patients with optic atrophy. Eur J Ophthalmol 2020; 31:3463-3470. [PMID: 33307808 DOI: 10.1177/1120672120977824] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Optic nerve cells can be irreversibly damaged by common various causes. Unfortunately optic nerve and retinal ganglion cells have no regenerative capacity and undergo apoptosis in case of damage. In this study, our aim is to investigate the safety and efficacy of suprachoroidal umbilical cord-derived MSCs (UC-MSCs) implantation in patients with optic atrophy. METHODS This study enrolled 29 eyes of 23 patients with optic atrophy who were followed in the ophthalmology department of our hospital. BCVA, anterior segment, fundus examination, color photography, and optical coherence tomography (OCT) were carried out at each visit. Fundus fluorescein angiography and visual field examination were performed at the end of the first, third, sixth months, and 1 year follow-up. RESULTS After suprachoroidal UC-MSCs implantation there were statistically significant improvements in BCVA and VF results during 12 months follow-up (p < 0.05). When we evaluate the results of VF tests, the mean deviation (MD) value at baseline was -26.11 ± 8.36 (range -14.18 to -34.41). At the end of the first year it improved to -25.01 ± 8.73 (range -12.56 to -34.41) which was statistically significant (p < 0.05). When we evaluate the mean RNFL thickness measurements at baseline and at 12 month follow-up the results were 81.8 ± 24.9 μm and 76.6 ± 22.6 μm, respectively. There was not a significant difference between the mean values (p > 0.05). CONCLUSION Stem cell treatment with suprachoroidal implantation of UCMSCs seems to be safe and effective in the treatment for optic nerve diseases that currently have no curative treatment options.
Collapse
Affiliation(s)
| | - Ayşe Öner
- Department of Ophthalmology, Acibadem Hospital, Kayseri, Turkey
| |
Collapse
|
11
|
Padhy SK, Takkar B, Narayanan R, Venkatesh P, Jalali S. Voretigene Neparvovec and Gene Therapy for Leber's Congenital Amaurosis: Review of Evidence to Date. APPLICATION OF CLINICAL GENETICS 2020; 13:179-208. [PMID: 33268999 PMCID: PMC7701157 DOI: 10.2147/tacg.s230720] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/06/2020] [Indexed: 12/13/2022]
Abstract
Gene therapy has now evolved as the upcoming modality for management of many disorders, both inheritable and non-inheritable. Knowledge of genetics pertaining to a disease has therefore become paramount for physicians across most specialities. Inheritable retinal dystrophies (IRDs) are notorious for progressive and relentless vision loss, frequently culminating in complete blindness in both eyes. Leber’s congenital amaurosis (LCA) is a typical example of an IRD that manifests very early in childhood. Research in gene therapy has led to the development and approval of voretigene neparvovec (VN) for use in patients of LCA with a deficient biallelic RPE65 gene. The procedure involves delivery of a recombinant virus vector that carries the RPE65 gene in the subretinal space. This comprehensive review reports the evidence thus far in support of gene therapy for LCA. We explore and compare the various gene targets including but not limited to RPE65, and discuss the choice of vector and method for ocular delivery. The review details the evolution of gene therapy with VN in a phased manner, concluding with the challenges that lie ahead for its translation for use in communities that differ much both genetically and economically.
Collapse
Affiliation(s)
- Srikanta Kumar Padhy
- Vitreoretina and Uveitis Services, L V Prasad Eye Institute, Mithu Tulsi Chanrai Campus, Bhubaneswar, India
| | - Brijesh Takkar
- Srimati Kanuri Santhamma Center for Vitreoretinal Diseases, Kallam Anji Reddy Campus, L.V. Prasad Eye Institute, Hyderabad, India.,Center of Excellence for Rare Eye Diseases, Kallam Anji Reddy Campus, L V Prasad Eye Institute, Hyderabad, India
| | - Raja Narayanan
- Srimati Kanuri Santhamma Center for Vitreoretinal Diseases, Kallam Anji Reddy Campus, L.V. Prasad Eye Institute, Hyderabad, India
| | - Pradeep Venkatesh
- Dr RP Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Subhadra Jalali
- Srimati Kanuri Santhamma Center for Vitreoretinal Diseases, Kallam Anji Reddy Campus, L.V. Prasad Eye Institute, Hyderabad, India.,Jasti V. Ramanamma Childrens' Eye Care Centre, Kallam Anji Reddy Campus, L V Prasad Eye Institute, Hyderabad, India
| |
Collapse
|
12
|
Mammadzada P, Corredoira PM, André H. The role of hypoxia-inducible factors in neovascular age-related macular degeneration: a gene therapy perspective. Cell Mol Life Sci 2020; 77:819-833. [PMID: 31893312 PMCID: PMC7058677 DOI: 10.1007/s00018-019-03422-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 12/04/2019] [Accepted: 12/10/2019] [Indexed: 12/19/2022]
Abstract
Understanding the mechanisms that underlie age-related macular degeneration (AMD) has led to the identification of key molecules. Hypoxia-inducible transcription factors (HIFs) have been associated with choroidal neovascularization and the progression of AMD into the neovascular clinical phenotype (nAMD). HIFs regulate the expression of multiple growth factors and cytokines involved in angiogenesis and inflammation, hallmarks of nAMD. This knowledge has propelled the development of a new group of therapeutic strategies focused on gene therapy. The present review provides an update on current gene therapies in ocular angiogenesis, particularly nAMD, from both basic and clinical perspectives.
Collapse
Affiliation(s)
- Parviz Mammadzada
- Division of Eye and Vision, Department of Clinical Neuroscience, Karolinska Institutet, St. Erik Eye Hospital, Stockholm, Sweden
| | - Pablo M Corredoira
- Division of Eye and Vision, Department of Clinical Neuroscience, Karolinska Institutet, St. Erik Eye Hospital, Stockholm, Sweden
| | - Helder André
- Division of Eye and Vision, Department of Clinical Neuroscience, Karolinska Institutet, St. Erik Eye Hospital, Stockholm, Sweden.
| |
Collapse
|
13
|
Doostmohammadi M, Forootanfar H, Ramakrishna S. Regenerative medicine and drug delivery: Progress via electrospun biomaterials. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 109:110521. [PMID: 32228899 DOI: 10.1016/j.msec.2019.110521] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 12/01/2019] [Accepted: 12/02/2019] [Indexed: 02/07/2023]
Abstract
Worldwide research on electrospinning enabled it as a versatile technique for producing nanofibers with specified physio-chemical characteristics suitable for diverse biomedical applications. In the case of tissue engineering and regenerative medicine, the nanofiber scaffolds' characteristics are custom designed based on the cells and tissues specific needs. This fabrication technique is also innovated for the production of nanofibers with special micro-structure and secondary structure characteristics such as porous fibers, hollow structure, and core- sheath structure. This review attempts to critically and succinctly capture the vast number of developments reported in the literature over the past two decades. We then discuss their applications as scaffolds for induction of cells growth and differentiation or as architecture for being used as graft for tissue engineering. The special nanofibers designed for improving regeneration of several tissues including heart, bone, central nerve system, spinal cord, skin and ocular tissue are introduced. We also discuss the potential of the electrospinning in drug delivery applications, which is a critical factor for cell culture, tissue formation and wound healing applications.
Collapse
Affiliation(s)
- Mohsen Doostmohammadi
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamid Forootanfar
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran; Herbal and Traditional Medicines Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Seeram Ramakrishna
- Department of Mechanical Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore.
| |
Collapse
|
14
|
Six-month results of suprachoroidal adipose tissue-derived mesenchymal stem cell implantation in patients with optic atrophy: a phase 1/2 study. Int Ophthalmol 2019; 39:2913-2922. [PMID: 31309439 DOI: 10.1007/s10792-019-01141-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 07/06/2019] [Indexed: 12/18/2022]
Abstract
PURPOSE This prospective clinical case series aimed to investigate the safety and efficacy of suprachoroidal adipose tissue-derived mesenchymal stem cell (ADMSC) implantation in patients with optic nerve diseases. METHODS This prospective, single-center, phase 1/2 study enrolled 4 eyes of 4 patients with optic atrophy of various reasons who underwent suprachoroidal implantation of ADMSCs. The best-corrected visual acuity (BCVA) in the study was HM at 1 m. The worse eye of the patient was operated. Patients were evaluated on the first day, first week, first month, third and sixth months postoperatively. BCVA, anterior segment and fundus examination, color photography, optical coherence tomography (OCT) and visual field examination were carried out at each visit. Fundus fluorescein angiography and multifocal electroretinography (mfERG) recordings were performed at the end of the first, third and sixth months and anytime if necessary during the follow-up. RESULTS All 4 patients completed the six-month follow-up. None of them had any systemic or ocular complications. All of the patients experienced visual acuity improvement, visual field improvement and improvement in the mfERG recordings. We found choroidal thickening in OCT of the 4 patients. CONCLUSION Even though the sample size is small, the improvements were still encouraging. Stem cell treatment with suprachoroidal implantation of ADMSCs seems to be safe and effective in the treatment for optic nerve diseases that currently have no curative treatment options.
Collapse
|
15
|
|
16
|
Kundu J, Michaelson A, Baranov P, Chiumiento M, Nigl T, Young MJ, Carrier RL. Interphotoreceptor matrix based biomaterial: Impact on human retinal progenitor cell attachment and differentiation. J Biomed Mater Res B Appl Biomater 2017; 106:891-899. [PMID: 28419733 DOI: 10.1002/jbm.b.33901] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 02/27/2017] [Accepted: 03/28/2017] [Indexed: 11/08/2022]
Abstract
While cell transplantation therapies show great promise as treatments for retinal degeneration, the challenge of low cell survival upon transplantation motivates exploration of materials that may serve as cell delivery vehicles and promote survival and differentiation. In this study, we explored the native matrix that surrounds the outer segments of photoreceptors and promotes their homeostasis, interphotoreceptor matrix (IPM), as a substrate for human retinal progenitor cells (hRPCs). Bovine IPM was characterized to determine its structure and biochemical composition, and processed to develop substrates for cells. Cell viability, morphology, proliferation and expression of photoreceptors marker genes were studied on IPM-based substrates in vitro. We explored different preparations of IPM as a scaffold. Lectin staining revealed that a distinct honeycomb structure of native IPM is lost during centrifugation to prepare a more concentrated suspension of matrix. Biochemical analysis of bovine IPM indicated presence of glycosaminoglycans and proteins. IPM mediated hRPC attachment and spreading with no signs of cytotoxicity. Cells proliferated more on native IPM substrates compared to IPM that was centrifuged to create a concentrated suspension. Cells cultured on IPM substrates expressed markers of photoreceptors: rhodopsin, NRL and ROM1. Together this data supports further exploration of IPM as a tool for retinal tissue engineering. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 891-899, 2018.
Collapse
Affiliation(s)
- Joydip Kundu
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, 02115.,Schepens Eye Research Institute, Massachusetts Eye and Ear, An affiliate of Harvard Medical School, Boston, Massachusetts, 02114
| | - Andrew Michaelson
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, 02115
| | - Petr Baranov
- Schepens Eye Research Institute, Massachusetts Eye and Ear, An affiliate of Harvard Medical School, Boston, Massachusetts, 02114
| | - Marco Chiumiento
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, 02115
| | - Tom Nigl
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, 02115
| | - Michael J Young
- Schepens Eye Research Institute, Massachusetts Eye and Ear, An affiliate of Harvard Medical School, Boston, Massachusetts, 02114
| | - Rebecca L Carrier
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, 02115
| |
Collapse
|
17
|
Correction of Pathological Morphofunctional Changes in the Mammalian Retina. NEUROPHYSIOLOGY+ 2016. [DOI: 10.1007/s11062-016-9549-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
18
|
Electrospun SF/PLCL nanofibrous membrane: a potential scaffold for retinal progenitor cell proliferation and differentiation. Sci Rep 2015; 5:14326. [PMID: 26395224 PMCID: PMC4585796 DOI: 10.1038/srep14326] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 08/24/2015] [Indexed: 12/22/2022] Open
Abstract
Biocompatible polymer scaffolds are promising as potential carriers for the delivery of retinal progenitor cells (RPCs) in cell replacement therapy for the repair of damaged or diseased retinas. The primary goal of the present study was to investigate the effects of blended electrospun nanofibrous membranes of silk fibroin (SF) and poly(L-lactic acid-co-ε-caprolactone) (PLCL), a novel scaffold, on the biological behaviour of RPCs in vitro. To assess the cell-scaffold interaction, RPCs were cultured on SF/PLCL scaffolds for indicated durations. Our data revealed that all the SF/PLCL scaffolds were thoroughly cytocompatible, and the SF:PLCL (1:1) scaffolds yielded the best RPC growth. The in vitro proliferation assays showed that RPCs proliferated more quickly on the SF:PLCL (1:1) than on the other scaffolds and the control. Quantitative polymerase chain reaction (qPCR) and immunocytochemistry analyses demonstrated that RPCs grown on the SF:PLCL (1:1) scaffolds preferentially differentiated toward retinal neurons, including, most interestingly, photoreceptors. In summary, we demonstrated that the SF:PLCL (1:1) scaffolds can not only markedly promote RPC proliferation with cytocompatibility for RPC growth but also robustly enhance RPCs’ differentiation toward specific retinal neurons of interest in vitro, suggesting that SF:PLCL (1:1) scaffolds may have potential applications in retinal cell replacement therapy in the future.
Collapse
|
19
|
Retinal synaptic regeneration via microfluidic guiding channels. Sci Rep 2015; 5:13591. [PMID: 26314276 PMCID: PMC4551994 DOI: 10.1038/srep13591] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 07/31/2015] [Indexed: 11/08/2022] Open
Abstract
In vitro culture of dissociated retinal neurons is an important model for investigating retinal synaptic regeneration (RSR) and exploring potentials in artificial retina. Here, retinal precursor cells were cultured in a microfluidic chip with multiple arrays of microchannels in order to reconstruct the retinal neuronal synapse. The cultured retinal cells were physically connected through microchannels. Activation of electric signal transduction by the cells through the microchannels was demonstrated by administration of glycinergic factors. In addition, an image-based analytical method was used to quantify the synaptic connections and to assess the kinetics of synaptic regeneration. The rate of RSR decreased significantly below 100 μM of inhibitor glycine and then approached to a relatively constant level at higher concentrations. Furthermore, RSR was enhanced by chemical stimulation with potassium chloride. Collectively, the microfluidic synaptic regeneration chip provides a novel tool for high-throughput investigation of RSR at the cellular level and may be useful in quality control of retinal precursor cell transplantation.
Collapse
|
20
|
Popelka Š, Studenovská H, Abelová L, Ardan T, Studený P, Straňák Z, Klíma J, Dvořánková B, Kotek J, Hodan J, Rypáček F. A frame-supported ultrathin electrospun polymer membrane for transplantation of retinal pigment epithelial cells. Biomed Mater 2015; 10:045022. [DOI: 10.1088/1748-6041/10/4/045022] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
21
|
Kvanta A, Grudzinska MK. Stem cell-based treatment in geographic atrophy: promises and pitfalls. Acta Ophthalmol 2014; 92:21-6. [PMID: 23890249 DOI: 10.1111/aos.12185] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Geographic atrophy is a common and untreatable form of advanced age-related macular degeneration. The degeneration primarily affects the retinal pigment epithelium and photoreceptors of the retina and their restoration by cell transplantation seems attractive. Recently, a patient with geographic atrophy was the first human to receive cells derived from human embryonic stem cells. In this short review, the rationale, potential and obstacles for stem cell-derived therapy in geographic atrophy are discussed.
Collapse
Affiliation(s)
- Anders Kvanta
- Department of Vitreoretinal Diseases, St. Erik Eye Hospital and Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
22
|
Haddad-Mashadrizeh A, Bahrami AR, Matin MM, Edalatmanesh MA, Zomorodipour A, Gardaneh M, Farshchian M, Momeni-Moghaddam M. Human adipose-derived mesenchymal stem cells can survive and integrate into the adult rat eye following xenotransplantation. Xenotransplantation 2013; 20:165-76. [PMID: 23679842 DOI: 10.1111/xen.12033] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 03/18/2013] [Indexed: 12/17/2022]
Abstract
BACKGROUND Novel threads of discovery provide the basis for optimism for the development of a stem-cell-based strategy for the treatment of retinal blindness. Accordingly, achievement to suitable cell source with potential-to-long-term survival and appropriate differentiation can be an effective step in this direction. METHODS After derivation of human adipose-derived mesenchymal stem cells (HAD-MSCs), they were stably transfected with a vector containing Turbo-green fluorescent protein (GFP) and JRed to be able to trace them after transplantation. Labeled HAD-MSCs were transplanted into the intact adult rat eye and their survival, integration, and migration during 6 months post-transplantation were assessed. RESULTS The transplanted cells were traceable in the rat vitreous humor (VH) up until 90 days after transplantation, with gradual reduction in numbers, their adhesion and expansion capacity after recovery. These cells were also integrated into the ocular tissues. Nonetheless, some of the implanted cells succeeded to cross the blood-retina barrier (BRB) and accumulate in the spleen with time. CONCLUSIONS The survival of the HAD-MSCs for a period of 90 days in VH and even longer period of up to 6 months in other eye tissues makes them a promising source to be considered in regenerative medicine of eye diseases. However, the potency of crossing the BRB by the implanted cells suggests that use of HAD-MSCs must be handled with extreme caution.
Collapse
Affiliation(s)
- Aliakbar Haddad-Mashadrizeh
- Cell and Molecular Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran; Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
The nervous system is characterized by its complex network of highly specialized cells that enable us to perceive stimuli from the outside world and react accordingly. The computational integration enabled by these networks remains to be elucidated, but appropriate sensory input, processing, and motor control are certainly essential for survival. Consequently, loss of nervous tissue due to injury or disease represents a considerable biomedical challenge. Stem cell research offers the promise to provide cells for nervous system repair to replace lost and damaged neural tissue and alleviate disease. We provide a protocol-based chapter on fundamental principles and procedures of pluripotent stem cell (PSC) differentiation and neural transplantation. Rather than detailed methodological step-by-step descriptions of these procedures, we provide an overview and highlight the most critical aspects and key steps of PSC neural induction, subtype specification in different in vitro systems, as well as neural cell transplantation to the central nervous system. We conclude with a summary of suitable readout methods including in vitro phenotypic analysis, histology, and functional analysis in vivo.
Collapse
|