1
|
Di Girolamo N. Biologicals and Biomaterials for Corneal Regeneration and Vision Restoration in Limbal Stem Cell Deficiency. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2401763. [PMID: 38777343 DOI: 10.1002/adma.202401763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/15/2024] [Indexed: 05/25/2024]
Abstract
The mammalian cornea is decorated with stem cells bestowed with the life-long task of renewing the epithelium, provided they remain healthy, functional, and in sufficient numbers. If not, a debilitating disease known as limbal stem cell deficiency (LSCD) can develop causing blindness. Decades after the first stem cell (SC) therapy is devised to treat this condition, patients continue to suffer unacceptable failures. During this time, improvements to therapeutics have included identifying better markers to isolate robust SC populations and nurturing them on crudely modified biological or biomaterial scaffolds including human amniotic membrane, fibrin, and contact lenses, prior to their delivery. Researchers are now gathering information about the biomolecular and biomechanical properties of the corneal SC niche to decipher what biological and/or synthetic materials can be incorporated into these carriers. Advances in biomedical engineering including electrospinning and 3D bioprinting with surface functionalization and micropatterning, and self-assembly models, have generated a wealth of biocompatible, biodegradable, integrating scaffolds to choose from, some of which are being tested for their SC delivery capacity in the hope of improving clinical outcomes for patients with LSCD.
Collapse
Affiliation(s)
- Nick Di Girolamo
- Mechanisms of Disease and Translational Research, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia
| |
Collapse
|
2
|
Lu C, Le Q. Advances in Organoid Technology: A Focus on Corneal Limbal Organoids. Stem Cell Rev Rep 2024; 20:1227-1235. [PMID: 38558362 DOI: 10.1007/s12015-024-10706-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2024] [Indexed: 04/04/2024]
Abstract
Organoid technology provides a versatile platform for simulating organogenesis, investigating disease pathogenesis, and exploring therapeutic interventions. Among various types of organoids that have been developed, corneal limbal organoids, the three-dimensional miniaturized corneas which are derived from either pluripotent stem cells or limbal epithelial stem cells, are particularly promising for clinical translation. This narrative review summarized the state-of-the-art in corneal limbal organoids research including the cultivation methods, clinical relevance and its limitations and challenges. The potential of corneal limbal organoids in mimicking corneal development, disease modelling, drug screening, and regenerative medicine was discussed. Technical improvements in cultivation techniques, imaging modalities, and gene editing tools are anticipated to overcome current limitations and further promote its clinical potential. Despite challenges and difficulties, the development of corneal limbal organoids opens a new era of regenerative medicine and provides a potential source of stem cell replacement therapies for challenging corneal diseases with the establishment of an in vitro corneal limbal organoid bank.
Collapse
Affiliation(s)
- Chuwei Lu
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, 200031, China
| | - Qihua Le
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, 200031, China.
- Research Center, Eye & ENT Hospital of Fudan University, Shanghai, 200031, China.
- Myopia Key Laboratory of Ministry of Health, Eye & ENT Hospital of Fudan University, Shanghai, 200031, China.
| |
Collapse
|
3
|
Koçak G, Uyulgan S, Polatlı E, Sarı V, Kahveci B, Bursali A, Binokay L, Reçber T, Nemutlu E, Mardinoğlu A, Karakülah G, Utine CA, Güven S. Generation of Anterior Segment of the Eye Cells from hiPSCs in Microfluidic Platforms. Adv Biol (Weinh) 2024; 8:e2400018. [PMID: 38640945 DOI: 10.1002/adbi.202400018] [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: 01/10/2024] [Revised: 03/10/2024] [Indexed: 04/21/2024]
Abstract
Ophthalmic diseases affect many people, causing partial or total loss of vision and a reduced quality of life. The anterior segment of the eye accounts for nearly half of all visual impairment that can lead to blindness. Therefore, there is a growing demand for ocular research and regenerative medicine that specifically targets the anterior segment to improve vision quality. This study aims to generate a microfluidic platform for investigating the formation of the anterior segment of the eye derived from human induced pluripotent stem cells (hiPSC) under various spatial-mechanoresponsive conditions. Microfluidic platforms are developed to examine the effects of dynamic conditions on the generation of hiPSCs-derived ocular organoids. The differentiation protocol is validated, and mechanoresponsive genes are identified through transcriptomic analysis. Several culture strategies is implemented for the anterior segment of eye cells in a microfluidic chip. hiPSC-derived cells showed anterior eye cell characteristics in mRNA and protein expression levels under dynamic culture conditions. The expression levels of yes-associated protein and transcriptional coactivator PDZ binding motif (YAP/TAZ) and PIEZO1, varied depending on the differentiation and growth conditions of the cells, as well as the metabolomic profiles under dynamic culture conditions.
Collapse
Affiliation(s)
- Gamze Koçak
- Izmir Biomedicine and Genome Center, Izmir, 35340, Türkiye
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, 35340, Türkiye
| | - Sude Uyulgan
- Izmir Biomedicine and Genome Center, Izmir, 35340, Türkiye
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, 35340, Türkiye
| | - Elifsu Polatlı
- Izmir Biomedicine and Genome Center, Izmir, 35340, Türkiye
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, 35340, Türkiye
| | - Vedat Sarı
- Izmir Biomedicine and Genome Center, Izmir, 35340, Türkiye
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, 35340, Türkiye
| | - Burak Kahveci
- Izmir Biomedicine and Genome Center, Izmir, 35340, Türkiye
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, 35340, Türkiye
| | - Ahmet Bursali
- Izmir Biomedicine and Genome Center, Izmir, 35340, Türkiye
| | - Leman Binokay
- Izmir Biomedicine and Genome Center, Izmir, 35340, Türkiye
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, 35340, Türkiye
| | - Tuba Reçber
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Sıhhiye, Ankara, 06100, Türkiye
| | - Emirhan Nemutlu
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Sıhhiye, Ankara, 06100, Türkiye
| | - Adil Mardinoğlu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, SE1 9RT, UK
| | - Gökhan Karakülah
- Izmir Biomedicine and Genome Center, Izmir, 35340, Türkiye
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, 35340, Türkiye
| | - Canan Aslı Utine
- Izmir Biomedicine and Genome Center, Izmir, 35340, Türkiye
- Department of Ophthalmology, Dokuz Eylül University Hospital, Dokuz Eylül University, Izmir, 35340, Türkiye
| | - Sinan Güven
- Izmir Biomedicine and Genome Center, Izmir, 35340, Türkiye
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, 35340, Türkiye
- Department of Medical Biology and Genetics, Faculty of Medicine, Dokuz Eylül University, Izmir, 35340, Türkiye
| |
Collapse
|
4
|
Patrício D, Santiago J, Mano JF, Fardilha M. Organoids of the male reproductive system: Challenges, opportunities, and their potential use in fertility research. WIREs Mech Dis 2023; 15:e1590. [PMID: 36442887 DOI: 10.1002/wsbm.1590] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/17/2022] [Accepted: 11/12/2022] [Indexed: 11/30/2022]
Abstract
Organoids are units of function of a given organ able to reproduce, in culture, a biological structure similar in architecture and function to its counterpart in vivo. Today, it is possible to develop an organoid from a fragment of tissue, a stem cell located in an adult organ, an embryonic stem cell, or an induced pluripotent stem cell. In the past decade, many organoids have been developed which mimic stomach, pancreas, liver and brain tissues, optic cups, among many others. Additionally, different male reproductive system organs have already been developed as organoids, including the prostate and testis. These 3D cultures may be of great importance for urological cancer research and have the potential to be used in fertility research for the study of spermatozoa production and maturation, germ cells-somatic cells interactions, and mechanisms of disease. They also provide an accurate preclinical pipeline for drug testing and discovery, as well as for the study of drug resistance. In this work, we revise the current knowledge on organoid technology and its use in healthcare and research, describe the male reproductive system organoids and other biomaterials already developed, and discuss their current application. Finally, we highlight the research gaps, challenges, and opportunities in the field and propose strategies to improve the use of organoids for the study of male infertility situations. This article is categorized under: Reproductive System Diseases > Stem Cells and Development Reproductive System Diseases > Biomedical Engineering.
Collapse
Affiliation(s)
- Daniela Patrício
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal.,Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Joana Santiago
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Margarida Fardilha
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
5
|
Asal M, Koçak G, Sarı V, Reçber T, Nemutlu E, Utine CA, Güven S. Development of lacrimal gland organoids from iPSC derived multizonal ocular cells. Front Cell Dev Biol 2023; 10:1058846. [PMID: 36684423 PMCID: PMC9846036 DOI: 10.3389/fcell.2022.1058846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/13/2022] [Indexed: 01/05/2023] Open
Abstract
Lacrimal gland plays a vital role in maintaining the health and function of the ocular surface. Dysfunction of the gland leads to disruption of ocular surface homeostasis and can lead to severe outcomes. Approaches evolving through regenerative medicine have recently gained importance to restore the function of the gland. Using human induced pluripotent stem cells (iPSCs), we generated functional in vitro lacrimal gland organoids by adopting the multi zonal ocular differentiation approach. We differentiated human iPSCs and confirmed commitment to neuro ectodermal lineage. Then we identified emergence of mesenchymal and epithelial lacrimal gland progenitor cells by the third week of differentiation. Differentiated progenitors underwent branching morphogenesis in the following weeks, typical of lacrimal gland development. We were able to confirm the presence of lacrimal gland specific acinar, ductal, and myoepithelial cells and structures during weeks 4-7. Further on, we demonstrated the role of miR-205 in regulation of the lacrimal gland organoid development by monitoring miR-205 and FGF10 mRNA levels throughout the differentiation process. In addition, we assessed the functionality of the organoids using the β-Hexosaminidase assay, confirming the secretory function of lacrimal organoids. Finally, metabolomics analysis revealed a shift from amino acid metabolism to lipid metabolism in differentiated organoids. These functional, tear proteins secreting human lacrimal gland organoids harbor a great potential for the improvement of existing treatment options of lacrimal gland dysfunction and can serve as a platform to study human lacrimal gland development and morphogenesis.
Collapse
Affiliation(s)
- Melis Asal
- Izmir Biomedicine and Genome Center, Izmir, Turkey,Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, Turkey
| | - Gamze Koçak
- Izmir Biomedicine and Genome Center, Izmir, Turkey,Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, Turkey
| | - Vedat Sarı
- Izmir Biomedicine and Genome Center, Izmir, Turkey,Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, Turkey
| | - Tuba Reçber
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Emirhan Nemutlu
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Canan Aslı Utine
- Izmir Biomedicine and Genome Center, Izmir, Turkey,Department of Ophthalmology, Dokuz Eylül University Hospital, Dokuz Eylül University, Izmir, Turkey
| | - Sinan Güven
- Izmir Biomedicine and Genome Center, Izmir, Turkey,Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, Turkey,Department of Medical Biology and Genetics, Faculty of Medicine, Dokuz Eylül University, Izmir, Turkey,*Correspondence: Sinan Güven,
| |
Collapse
|
6
|
The progress in techniques for culturing human limbal epithelial stem cells. Hum Cell 2023; 36:1-14. [PMID: 36181663 DOI: 10.1007/s13577-022-00794-2] [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: 07/21/2022] [Accepted: 09/11/2022] [Indexed: 01/07/2023]
Abstract
In vitro culture of human limbal epithelial stem cells (hLESCs) is crucial to cell therapy in the treatment of limbal stem cell deficiency, a potentially vision-threatening disease that is characterized by persistent corneal epithelial defects and corneal epithelium conjunctivalization. Traditionally, hLESCs are cultivated based on either limbal tissue explants or single-cell suspensions in culture media containing xenogenous components, such as fetal bovine serum and murine 3T3 feeder cells. Plastic culture dishes and human amniotic membranes are classical growth substrates used in conventional hLESC culture systems. The past few decades have witnessed considerable progress and innovations in hLESC culture techniques to ensure a higher level of biosafety and lower immunogenicity for further cell treatment, including complete removal of xenogenous components from culture media, the application of human-derived feeder cells, and the development of novel scaffolds. Three-dimensional artificial niches and three-dimensional culture techniques have also been established to simulate the real microenvironment of limbal crypts for better cell outgrowth and proliferation. All these progresses ensure that in vitro cultured hLESCs are more adaptable to translational stem cell therapy for limbal stem cell deficiency.
Collapse
|
7
|
Miotti G, Parodi PC, Ferrari A, Salati C, Zeppieri M. Stem Cells in Ophthalmology: From the Bench to the Bedside. HANDBOOK OF STEM CELL APPLICATIONS 2023:1-24. [DOI: 10.1007/978-981-99-0846-2_10-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/22/2023] [Indexed: 09/13/2023]
|
8
|
Miotti G, Parodi PC, Ferrari A, Salati C, Zeppieri M. Stem Cells in Ophthalmology: From the Bench to the Bedside. HANDBOOK OF STEM CELL APPLICATIONS 2023:1-24. [DOI: https:/doi.org/10.1007/978-981-99-0846-2_10-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/22/2023] [Indexed: 08/28/2023]
|
9
|
Sullivan KM, Ko E, Kim EM, Ballance WC, Ito JD, Chalifoux M, Kim YJ, Bashir R, Kong H. Extracellular Microenvironmental Control for Organoid Assembly. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:1209-1222. [PMID: 35451330 PMCID: PMC9836674 DOI: 10.1089/ten.teb.2021.0186] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/04/2022] [Indexed: 01/22/2023]
Abstract
Organoids, which are multicellular clusters with similar physiological functions to living organs, have gained increasing attention in bioengineering. As organoids become more advanced, methods to form complex structures continue to develop. There is evidence that the extracellular microenvironment can regulate organoid quality. The extracellular microenvironment consists of soluble bioactive molecules, extracellular matrix, and biofluid flow. However, few efforts have been made to discuss the microenvironment optimal to engineer specific organoids. Therefore, this review article examines the extent to which engineered extracellular microenvironments regulate organoid quality. First, we summarize the natural tissue and organ's unique chemical and mechanical properties, guiding researchers to design an extracellular microenvironment used for organoid engineering. Then, we summarize how the microenvironments contribute to the formation and growth of the brain, lung, intestine, liver, retinal, and kidney organoids. The approaches to forming and evaluating the resulting organoids are also discussed in detail. Impact statement Organoids, which are multicellular clusters with similar physiological function to living organs, have been gaining increasing attention in bioengineering. As organoids become more advanced, methods to form complex structures continue to develop. This review article focuses on recent efforts to engineer the extracellular microenvironment in organoid research. We summarized the natural organ's microenvironment, which informs researchers of key factors that can influence organoid formation. Then, we summarize how these microenvironmental controls significantly contribute to the formation and growth of the corresponding brain, lung, intestine, liver, retinal, and kidney organoids. The approaches to forming and evaluating the resulting organoids are discussed in detail, including extracellular matrix choice and properties, culture methods, and the evaluation of the morphology and functionality through imaging and biochemical analysis.
Collapse
Affiliation(s)
- Kathryn M. Sullivan
- Department of Bioengineering, and University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| | - Eunkyung Ko
- Department of Bioengineering, and University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| | - Eun Mi Kim
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| | - William C. Ballance
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| | - John D. Ito
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| | - Madeleine Chalifoux
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| | - Young Jun Kim
- Environmental Safety Group, Korea Institute of Science and Technology (KIST–Europe), Saarbrucken, Germany
| | - Rashid Bashir
- Department of Bioengineering, and University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| | - Hyunjoon Kong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana−Champaign, Urbana, Illinois, USA
| |
Collapse
|
10
|
Ying PX, Fu M, Huang C, Li ZH, Mao QY, Fu S, Jia XH, Cao YC, Hong LB, Cai LY, Guo X, Liu RB, Meng FK, Yi GG. Profile of biological characterizations and clinical application of corneal stem/progenitor cells. World J Stem Cells 2022; 14:777-797. [PMID: 36483848 PMCID: PMC9724387 DOI: 10.4252/wjsc.v14.i11.777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/08/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
Corneal stem/progenitor cells are typical adult stem/progenitor cells. The human cornea covers the front of the eyeball, which protects the eye from the outside environment while allowing vision. The location and function demand the cornea to maintain its transparency and to continuously renew its epithelial surface by replacing injured or aged cells through a rapid turnover process in which corneal stem/progenitor cells play an important role. Corneal stem/progenitor cells include mainly corneal epithelial stem cells, corneal endothelial cell progenitors and corneal stromal stem cells. Since the discovery of corneal epithelial stem cells (also known as limbal stem cells) in 1971, an increasing number of markers for corneal stem/progenitor cells have been proposed, but there is no consensus regarding the definitive markers for them. Therefore, the identification, isolation and cultivation of these cells remain challenging without a unified approach. In this review, we systematically introduce the profile of biological characterizations, such as anatomy, characteristics, isolation, cultivation and molecular markers, and clinical applications of the three categories of corneal stem/progenitor cells.
Collapse
Affiliation(s)
- Pei-Xi Ying
- Department of Ophthalmology, Zhujiang Hospital, The Second Clinical School, Southern Medical University, Guangzhou 510280, Guangdong Province, China
| | - Min Fu
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, China
| | - Chang Huang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200030, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai 200030, China
- Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200030, China
| | - Zhi-Hong Li
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Lab of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510550, Guangdong Province, China
| | - Qing-Yi Mao
- The Second Clinical School, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Sheng Fu
- Hengyang Medical School, The University of South China, Hengyang 421001, Hunan Province, China
| | - Xu-Hui Jia
- The Second Clinical School, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Yu-Chen Cao
- The Second Clinical School, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Li-Bing Hong
- The Second Clinical School, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Li-Yang Cai
- The Second Clinical School, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Xi Guo
- Medical College of Rehabilitation, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Ru-Bing Liu
- The Second Clinical School, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Fan-ke Meng
- Emergency Department, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, China
| | - Guo-Guo Yi
- Department of Ophthalmology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
| |
Collapse
|
11
|
Bedos L, Wickham H, Gabriel V, Zdyrski C, Allbaugh RA, Sahoo DK, Sebbag L, Mochel JP, Allenspach K. Culture and characterization of canine and feline corneal epithelial organoids: A new tool for the study and treatment of corneal diseases. Front Vet Sci 2022; 9:1050467. [PMID: 36406087 PMCID: PMC9672346 DOI: 10.3389/fvets.2022.1050467] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
In this study, we isolated and cultured canine and feline 3D corneal organoids. Samples derived from corneal limbal epithelium from one canine and one feline patient were obtained by enucleation after euthanasia. Stem cell isolation and organoid culture were performed by culturing organoids in Matrigel. Organoids were subsequently embedded in paraffin for further characterization. The expression of key corneal epithelial and stromal cell markers in canine and feline organoids was evaluated at the mRNA level by RNA-ISH and at the protein level by immunofluorescence (IF) and immunohistochemistry (IHC), while histochemical analysis was performed on both tissues and organoids using periodic-acid Schiff (PAS), Sirius Red, Gomori's Trichrome, and Colloidal Iron stains. IF showed consistent expression of AQP1 within canine and feline organoids and tissues. P63 was present in canine tissues, canine organoids, and feline tissues, but not in feline organoids. Results from IHC staining further confirmed the primarily epithelial origin of the organoids. Canine and feline 3D corneal organoids can successfully be cultured and maintained and express epithelial and stem cell progenitor markers typical of the cornea. This novel in vitro model can be used in veterinary ophthalmology disease modeling, corneal drug testing, and regenerative medicine.
Collapse
Affiliation(s)
- Leila Bedos
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States
| | - Hannah Wickham
- SMART Lab, Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
| | - Vojtech Gabriel
- SMART Lab, Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
| | - Christopher Zdyrski
- SMART Lab, Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
| | - Rachel A. Allbaugh
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States
| | - Dipak Kumar Sahoo
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States
| | - Lionel Sebbag
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States
- Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Jonathan P. Mochel
- SMART Lab, Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
- 3D Health Solutions Inc., Ames, IA, United States
| | - Karin Allenspach
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States
- SMART Lab, Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
- 3D Health Solutions Inc., Ames, IA, United States
- *Correspondence: Karin Allenspach
| |
Collapse
|
12
|
Keng CT, Guo K, Liu YC, Shen KY, Lim DS, Lovatt M, Ang HP, Mehta JS, Chew WL. Multiplex viral tropism assay in complex cell populations with single-cell resolution. Gene Ther 2022; 29:555-565. [PMID: 35999303 PMCID: PMC9482877 DOI: 10.1038/s41434-022-00360-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 06/23/2022] [Accepted: 07/12/2022] [Indexed: 11/09/2022]
Abstract
Gene therapy constitutes one of the most promising mode of disease treatments. Two key properties for therapeutic delivery vectors are its transduction efficiency (how well the vector delivers therapeutic cargo to desired target cells) and specificity (how well it avoids off-target delivery into unintended cells within the body). Here we developed an integrated bioinformatics and experimental pipeline that enables multiplex measurement of transduction efficiency and specificity, particularly by measuring how libraries of delivery vectors transduce libraries of diverse cell types. We demonstrated that pairing high-throughput measurement of AAV identity with high-resolution single-cell RNA transcriptomic sequencing maps how natural and engineered AAV variants transduce individual cells within human cerebral and ocular organoids. We further demonstrate that efficient AAV transduction observed in organoids is recapitulated in vivo in non-human primates. This library-on-library technology will be important for determining the safety and efficacy of therapeutic delivery vectors.
Collapse
Affiliation(s)
- Choong Tat Keng
- Genome Institute of Singapore, Agency for Science, Technology and Research, 60 Biopolis Street, Singapore, 138672, Singapore
| | - Ke Guo
- Genome Institute of Singapore, Agency for Science, Technology and Research, 60 Biopolis Street, Singapore, 138672, Singapore
| | - Yu-Chi Liu
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore, Singapore.,Cornea and Refractive Surgery Group, Singapore Eye Research Institute, Singapore, Singapore.,Cornea and External Eye Diseases, Singapore National Eye Centre, Singapore, Singapore.,Ophthalmology Academic Clinical Program, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Kimberle Yanyin Shen
- Genome Institute of Singapore, Agency for Science, Technology and Research, 60 Biopolis Street, Singapore, 138672, Singapore
| | - Daryl Shern Lim
- Genome Institute of Singapore, Agency for Science, Technology and Research, 60 Biopolis Street, Singapore, 138672, Singapore
| | - Matthew Lovatt
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore, Singapore
| | - Heng Pei Ang
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore, Singapore
| | - Jodhbir S Mehta
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore, Singapore.,Cornea and Refractive Surgery Group, Singapore Eye Research Institute, Singapore, Singapore.,Cornea and External Eye Diseases, Singapore National Eye Centre, Singapore, Singapore.,Ophthalmology Academic Clinical Program, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Wei Leong Chew
- Genome Institute of Singapore, Agency for Science, Technology and Research, 60 Biopolis Street, Singapore, 138672, Singapore. .,Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore.
| |
Collapse
|
13
|
Cultured Autologous Corneal Epithelia for the Treatment of Unilateral Limbal Stem Cell Deficiency: A Case Series of 15 Patients. Biomedicines 2022; 10:biomedicines10081958. [PMID: 36009509 PMCID: PMC9405734 DOI: 10.3390/biomedicines10081958] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/01/2022] [Accepted: 08/08/2022] [Indexed: 11/17/2022] Open
Abstract
Damage to limbal epithelial stem cells can lead to limbal stem cell deficiency (LSCD). Current autologous treatment procedures for unilateral LSCD bear a significant risk of inducing LSCD in the donor eye. This complication can be avoided by grafting a stem cell containing cultured autologous corneal epithelium (CACE). The primary objective of this study was to demonstrate the safety of CACE grafted on eyes with LSCD. The secondary objective was to assess the efficacy of a CACE graft in restoring a self-renewing corneal surface with adequate anatomic structures, as well as improving the best corrected visual acuity (BCVA). Fifteen patients were grafted with a CACE on a fibrin gel produced from a 3 mm2 limbal biopsy harvested from the donor eye. Data were collected at baseline and after grafting. Follow-ups from 1 to 5 years were conducted. No major adverse events related to the CACE graft were observed. For every visit, an anatomic score based on corneal opacity as well as central vascularization and a functional score based on BCVA were determined. Safety was demonstrated by the low occurrence of complications. Anatomical (93%) and functional (47%) results are promising for improving vision in LSCD patients. Combined functional success and partial success rates with inclusion of BCVA were 53% [CI95: 27–79%] one year after CACE grafting. At the last follow-up, 87% [CI95: 60–98%] of the patients had attained corneal clarity. The outcomes demonstrate the safety of our technique and are promising regarding the efficacy of CACE in these patients.
Collapse
|
14
|
Santra M, Liu YC, Jhanji V, Yam GHF. Human SMILE-Derived Stromal Lenticule Scaffold for Regenerative Therapy: Review and Perspectives. Int J Mol Sci 2022; 23:ijms23147967. [PMID: 35887309 PMCID: PMC9315730 DOI: 10.3390/ijms23147967] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/10/2022] [Accepted: 07/18/2022] [Indexed: 12/13/2022] Open
Abstract
A transparent cornea is paramount for vision. Corneal opacity is one of the leading causes of blindness. Although conventional corneal transplantation has been successful in recovering patients’ vision, the outcomes are challenged by a global lack of donor tissue availability. Bioengineered corneal tissues are gaining momentum as a new source for corneal wound healing and scar management. Extracellular matrix (ECM)-scaffold-based engineering offers a new perspective on corneal regenerative medicine. Ultrathin stromal laminar tissues obtained from lenticule-based refractive correction procedures, such as SMall Incision Lenticule Extraction (SMILE), are an accessible and novel source of collagen-rich ECM scaffolds with high mechanical strength, biocompatibility, and transparency. After customization (including decellularization), these lenticules can serve as an acellular scaffold niche to repopulate cells, including stromal keratocytes and stem cells, with functional phenotypes. The intrastromal transplantation of these cell/tissue composites can regenerate native-like corneal stromal tissue and restore corneal transparency. This review highlights the current status of ECM-scaffold-based engineering with cells, along with the development of drug and growth factor delivery systems, and elucidates the potential uses of stromal lenticule scaffolds in regenerative therapeutics.
Collapse
Affiliation(s)
- Mithun Santra
- Corneal Regeneration Laboratory, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (M.S.); (V.J.)
| | - Yu-Chi Liu
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore;
- Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Vishal Jhanji
- Corneal Regeneration Laboratory, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (M.S.); (V.J.)
| | - Gary Hin-Fai Yam
- Corneal Regeneration Laboratory, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (M.S.); (V.J.)
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore;
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Correspondence:
| |
Collapse
|
15
|
Tafti MF, Aghamollaei H, Moghaddam MM, Jadidi K, Alio JL, Faghihi S. Emerging tissue engineering strategies for the corneal regeneration. J Tissue Eng Regen Med 2022; 16:683-706. [PMID: 35585479 DOI: 10.1002/term.3309] [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: 11/23/2021] [Revised: 04/16/2022] [Accepted: 04/19/2022] [Indexed: 11/10/2022]
Abstract
Cornea as the outermost layer of the eye is at risk of various genetic and environmental diseases that can damage the cornea and impair vision. Corneal transplantation is among the most applicable surgical procedures for repairing the defected tissue. However, the scarcity of healthy tissue donations as well as transplantation failure has remained as the biggest challenges in confront of corneal grafting. Therefore, alternative approaches based on stem-cell transplantation and classic regenerative medicine have been developed for corneal regeneration. In this review, the application and limitation of the recently-used advanced approaches for regeneration of cornea are discussed. Additionally, other emerging powerful techniques such as 5D printing as a new branch of scaffold-based technologies for construction of tissues other than the cornea are highlighted and suggested as alternatives for corneal reconstruction. The introduced novel techniques may have great potential for clinical applications in corneal repair including disease modeling, 3D pattern scheming, and personalized medicine.
Collapse
Affiliation(s)
- Mahsa Fallah Tafti
- Stem Cell and Regenerative Medicine Group, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Hossein Aghamollaei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Khosrow Jadidi
- Vision Health Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Jorge L Alio
- Department of Research and Development, VISSUM, Alicante, Spain.,Cornea, Cataract and Refractive Surgery Department, VISSUM, Alicante, Spain.,Department of Pathology and Surgery, Division of Ophthalmology, Faculty of Medicine, Miguel Hernández University, Alicante, Spain
| | - Shahab Faghihi
- Stem Cell and Regenerative Medicine Group, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
16
|
da Mata Martins TM, de Carvalho JL, da Silva Cunha P, Gomes DA, de Goes AM. Induction of Corneal Epithelial Differentiation of Induced Pluripotent and Orbital Fat-Derived Stem Cells Seeded on Decellularized Human Corneas. Stem Cell Rev Rep 2022; 18:2522-2534. [PMID: 35247143 DOI: 10.1007/s12015-022-10356-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2022] [Indexed: 11/28/2022]
Abstract
Up to 40% of donor corneas are deemed unsuitable for transplantation, aggravating the shortage of graft tissue. In most cases, the corneal extracellular matrix is intact. Therefore, their decellularization followed by repopulation with autologous cells may constitute an efficient alternative to reduce the amount of discarded tissue and the risk of immune rejection after transplantation. Although induced pluripotent (hiPSCs) and orbital fat-derived stem cells (OFSCs) hold great promise for corneal epithelial (CE) reconstruction, no study to date has evaluated the capacity of decellularized corneas (DCs) to support the attachment and differentiation of these cells into CE-like cells. Here, we recellularize DCs with hiPSCs and OFSCs and evaluate their differentiation potential into CE-like cells using animal serum-free culture conditions. Cell viability and adhesion on DCs were assessed by calcein-AM staining and scanning electron microscopy. Cell differentiation was evaluated by RT-qPCR and immunofluorescence analyses. DCs successfully supported the adhesion and survival of hiPSCs and OFSCs. The OFSCs cultured under differentiation conditions could not express the CE markers, TP63, KRT3, PAX6, and KRT12, while the hiPSCs gave rise to cells expressing high levels of these markers. RT-qPCR data suggested that the DCs provided an inductive environment for CE differentiation of hiPSCs, supporting the expression of PAX6 and KRT12 without the need for any soluble induction factors. Our results open the avenue for future studies regarding the in vivo effects of DCs as carriers for autologous cell transplantation for ocular surface reconstruction.
Collapse
Affiliation(s)
- Thaís Maria da Mata Martins
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte, Minas Gerais, 31270-901, Brazil.
| | - Juliana Lott de Carvalho
- Department of Genomic Sciences and Biotechnology, Catholic University of Brasilia, QS 07 - Lote 01, EPCT - Taguatinga, Brasília, Distrito Federal, 71966-700, Brazil.,Faculty of Medicine, University of Brasilia, Campus Universitário Darcy Ribeiro, Brasília, Distrito Federal, 70910-900, Brazil
| | - Pricila da Silva Cunha
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte, Minas Gerais, 31270-901, Brazil.,Department of Biology, Minas Gerais State University, Avenida Olegário Maciel, 1427, Ubá, Minas Gerais, 36502-002, Brazil
| | - Dawidson Assis Gomes
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Alfredo Miranda de Goes
- Department of Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| |
Collapse
|
17
|
Isla-Magrané H, Veiga A, García-Arumí J, Duarri A. Multiocular organoids from human induced pluripotent stem cells displayed retinal, corneal, and retinal pigment epithelium lineages. Stem Cell Res Ther 2021; 12:581. [PMID: 34809716 PMCID: PMC8607587 DOI: 10.1186/s13287-021-02651-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/05/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Recently, great efforts have been made to design protocols for obtaining ocular cells from human stem cells to model diseases or for regenerative purposes. Current protocols generally focus on isolating retinal cells, retinal pigment epithelium (RPE), or corneal cells and fail to recapitulate the complexity of the tissue during eye development. Here, the generation of more advanced in vitro multiocular organoids from human induced pluripotent stem cells (hiPSCs) is demonstrated. METHODS A 2-step method was established to first obtain self-organized multizone ocular progenitor cells (mzOPCs) from 2D hiPSC cultures within three weeks. Then, after the cells were manually isolated and grown in suspension, 3D multiocular organoids were generated to model important cellular features of developing eyes. RESULTS In the 2D culture, self-formed mzOPCs spanned the neuroectoderm, surface ectoderm, neural crest, and RPE, mimicking early stages of eye development. After lifting, mzOPCs developed into different 3D multiocular organoids composed of multiple cell lineages including RPE, retina, and cornea, and interactions between the different cell types and regions of the eye system were observed. Within these organoids, the retinal regions exhibited correct layering and contained all major retinal cell subtypes as well as retinal morphological cues, whereas the corneal regions closely resembled the transparent ocular-surface epithelium and contained of corneal, limbal, and conjunctival epithelial cells. The arrangement of RPE cells also formed organoids composed of polarized pigmented epithelial cells at the surface that were completely filled with collagen matrix. CONCLUSIONS This approach clearly demonstrated the advantages of the combined 2D-3D construction tissue model as it provided a more ocular native-like cellular environment than that of previous models. In this complex preparations, multiocular organoids may be used to model the crosstalk between different cell types in eye development and disease.
Collapse
Affiliation(s)
- Helena Isla-Magrané
- Ophthalmology Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Anna Veiga
- Regenerative Medicine Program IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - José García-Arumí
- Ophthalmology Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Department of Ophthalmology, Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Department of Ophthalmology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Anna Duarri
- Ophthalmology Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain.
| |
Collapse
|
18
|
Mijanović O, Pylaev T, Nikitkina A, Artyukhova M, Branković A, Peshkova M, Bikmulina P, Turk B, Bolevich S, Avetisov S, Timashev P. Tissue Engineering Meets Nanotechnology: Molecular Mechanism Modulations in Cornea Regeneration. MICROMACHINES 2021; 12:mi12111336. [PMID: 34832752 PMCID: PMC8618371 DOI: 10.3390/mi12111336] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 09/23/2021] [Accepted: 10/14/2021] [Indexed: 12/13/2022]
Abstract
Nowadays, tissue engineering is one of the most promising approaches for the regeneration of various tissues and organs, including the cornea. However, the inability of biomaterial scaffolds to successfully integrate into the environment of surrounding tissues is one of the main challenges that sufficiently limits the restoration of damaged corneal tissues. Thus, the modulation of molecular and cellular mechanisms is important and necessary for successful graft integration and long-term survival. The dynamics of molecular interactions affecting the site of injury will determine the corneal transplantation efficacy and the post-surgery clinical outcome. The interactions between biomaterial surfaces, cells and their microenvironment can regulate cell behavior and alter their physiology and signaling pathways. Nanotechnology is an advantageous tool for the current understanding, coordination, and directed regulation of molecular cell-transplant interactions on behalf of the healing of corneal wounds. Therefore, the use of various nanotechnological strategies will provide new solutions to the problem of corneal allograft rejection, by modulating and regulating host-graft interaction dynamics towards proper integration and long-term functionality of the transplant.
Collapse
Affiliation(s)
- Olja Mijanović
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia; (A.N.); (M.A.); (M.P.); (P.B.); (B.T.); (P.T.)
- Correspondence:
| | - Timofey Pylaev
- Saratov Medical State University N.A. V.I. Razumovsky, 112 Bolshaya Kazachya St., 410012 Saratov, Russia;
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences, 13 Prospekt Entuziastov, 410049 Saratov, Russia
| | - Angelina Nikitkina
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia; (A.N.); (M.A.); (M.P.); (P.B.); (B.T.); (P.T.)
| | - Margarita Artyukhova
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia; (A.N.); (M.A.); (M.P.); (P.B.); (B.T.); (P.T.)
| | - Ana Branković
- Department of Forensic Engineering, University of Criminal Investigation and Police Studies, 196 Cara Dušana St., Belgrade 11000, Serbia;
| | - Maria Peshkova
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia; (A.N.); (M.A.); (M.P.); (P.B.); (B.T.); (P.T.)
- World-Class Research Center “Digital biodesign and personalized healthcare”, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia
| | - Polina Bikmulina
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia; (A.N.); (M.A.); (M.P.); (P.B.); (B.T.); (P.T.)
- World-Class Research Center “Digital biodesign and personalized healthcare”, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia
| | - Boris Turk
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia; (A.N.); (M.A.); (M.P.); (P.B.); (B.T.); (P.T.)
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Sergey Bolevich
- Department of Human Pathology, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia;
| | - Sergei Avetisov
- Department of Eye Diseases, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia;
- Research Institute of Eye Diseases, 11 Rossolimo St., 119021 Moscow, Russia
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia; (A.N.); (M.A.); (M.P.); (P.B.); (B.T.); (P.T.)
- World-Class Research Center “Digital biodesign and personalized healthcare”, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, Leninskiye Gory 1-3, 119991 Moscow, Russia
| |
Collapse
|
19
|
Bonneau N, Baudouin C, Réaux-Le Goazigo A, Brignole-Baudouin F. An overview of current alternative models in the context of ocular surface toxicity. J Appl Toxicol 2021; 42:718-737. [PMID: 34648674 DOI: 10.1002/jat.4246] [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: 06/11/2021] [Revised: 08/31/2021] [Accepted: 09/15/2021] [Indexed: 11/06/2022]
Abstract
The 21st century has seen a steadily increasing social awareness of animal suffering, with increased attention to ethical considerations. Developing new integrated approaches to testing and assessment (IATA) strategies is an Organisation for Economic Co-operation and Development (OECD) goal to reduce animal testing. Currently, there is a lack of alternative models to test for ocular surface toxicity (aside from irritation) in lieu of the Draize eye irritation test (OECD guideline No. 405) performed in rabbits. Five alternative in vitro or ex vivo methods have been validated to replace this reference test, but only in combination. However, pathologies like Toxicity-Induced Dry Eye (TIDE), cataract, glaucoma, and neuropathic pain can occur after exposure to a pharmaceutical product or chemical and therefore need to be anticipated. To do so, new models of lacrimal glands, lens, and neurons innervating epithelia are required. These models must take into account real-life exposure (dose, time, and tear film clearance). The scientific community is working hard to develop new, robust, alternative, in silico, and in vitro models, while attempting to balance ethics and availability of biological materials. This review provides a broad overview of the validated methods for analyzing ocular irritation and those still used by some industries, as well as promising models that need to be optimized according to the OECD. Finally, we give an overview of recently developed innovative models, which could become new tools in the evaluation of ocular surface toxicity within the scope of IATAs.
Collapse
Affiliation(s)
- Noémie Bonneau
- Sorbonne Université, INSERM, CNRS, IHU FOReSight, Institut de la Vision, Paris, France.,Horus Pharma, Saint-Laurent-du-Var, France
| | - Christophe Baudouin
- Sorbonne Université, INSERM, CNRS, IHU FOReSight, Institut de la Vision, Paris, France.,Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DGOS CIC 1423, IHU FOReSight, Paris, France.,Université Versailles-Saint-Quentin-en-Yvelines, Hôpital Ambroise Paré, APHP, Boulogne-Billancourt, France
| | | | - Françoise Brignole-Baudouin
- Sorbonne Université, INSERM, CNRS, IHU FOReSight, Institut de la Vision, Paris, France.,Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DGOS CIC 1423, IHU FOReSight, Paris, France.,Laboratoire d'Ophtalmobiologie, Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, IHU FOReSight, Paris, France.,Université de Paris, Faculté de Pharmacie de Paris, Département de Toxicologie, Paris, France
| |
Collapse
|
20
|
Complex Organ Construction from Human Pluripotent Stem Cells for Biological Research and Disease Modeling with New Emerging Techniques. Int J Mol Sci 2021; 22:ijms221910184. [PMID: 34638524 PMCID: PMC8508560 DOI: 10.3390/ijms221910184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 12/13/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) are grouped into two cell types; embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs). hESCs have provided multiple powerful platforms to study human biology, including human development and diseases; however, there were difficulties in the establishment of hESCs from human embryo and concerns over its ethical issues. The discovery of hiPSCs has expanded to various applications in no time because hiPSCs had already overcome these problems. Many hPSC-based studies have been performed using two-dimensional monocellular culture methods at the cellular level. However, in many physiological and pathophysiological conditions, intra- and inter-organ interactions play an essential role, which has hampered the establishment of an appropriate study model. Therefore, the application of recently developed technologies, such as three-dimensional organoids, bioengineering, and organ-on-a-chip technology, has great potential for constructing multicellular tissues, generating the functional organs from hPSCs, and recapitulating complex tissue functions for better biological research and disease modeling. Moreover, emerging techniques, such as single-cell transcriptomics, spatial transcriptomics, and artificial intelligence (AI) allowed for a denser and more precise analysis of such heterogeneous and complex tissues. Here, we review the applications of hPSCs to construct complex organs and discuss further prospects of disease modeling and drug discovery based on these PSC-derived organs.
Collapse
|
21
|
Singh V, Tiwari A, Kethiri AR, Sangwan VS. Current perspectives of limbal-derived stem cells and its application in ocular surface regeneration and limbal stem cell transplantation. Stem Cells Transl Med 2021; 10:1121-1128. [PMID: 33951336 PMCID: PMC8284782 DOI: 10.1002/sctm.20-0408] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 02/11/2021] [Accepted: 03/04/2021] [Indexed: 12/13/2022] Open
Abstract
Limbal stem cells are involved in replenishing and maintaining the epithelium of the cornea. Damage to the limbus due to chemical/physical injury, infections, or genetic disorders leads to limbal stem cell deficiency (LSCD) with partial or total vision loss. Presently, LSCD is treated by transplanting limbal stem cells from the healthy eye of the recipient, living-related, or cadaveric donors. This review discusses limbal-derived stem cells, the importance of extracellular matrix in stem cell niche maintenance, the historical perspective of treating LSCD, including related advantages and limitations, and our experience of limbal stem cell transplantation over the decades.
Collapse
Affiliation(s)
- Vivek Singh
- Stem Cell Biology Laboratory, Center for Ocular Regeneration, L V Prasad Eye Institute, Hyderabad, India
| | - Anil Tiwari
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Cornea and Uveitis, Dr. Shroff's Charity Eye Hospital, New Delhi, India
| | - Abhinav Reddy Kethiri
- Stem Cell Biology Laboratory, Center for Ocular Regeneration, L V Prasad Eye Institute, Hyderabad, India
| | | |
Collapse
|
22
|
Van Meenen J, Ní Dhubhghaill S, Van den Bogerd B, Koppen C. An Overview of Advanced In Vitro Corneal Models: Implications for Pharmacological Testing. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:506-516. [PMID: 33878935 DOI: 10.1089/ten.teb.2021.0031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The cornea is an important barrier to consider when developing ophthalmic formulations, but proper modeling of this multilayered tissue remains a challenge. This is due to the varying properties associated with each layer in addition to the dynamics of the tear film. Hence, the most representative models to date rely on animals. Animal models, however, differ from humans in several aspects and are subject to ethical limitations. Consequently, in vitro approaches are being developed to address these issues. This review focuses on the barrier properties of the cornea and evaluates the most advanced three-dimensional cultures of human corneal equivalents in literature. Their application potential is subsequently assessed and discussed in the context of preclinical testing along with our perspective toward the future. Impact statement Most ocular drugs are applied topically, with the transcorneal pathway as the main administration route. Animal corneas are currently the only advanced models available, contributing to the drug attrition rate. Anatomical and physiological interspecies differences might account for a poor translatability of preclinical results to clinical trials, urging researchers to devise better corneal equivalents. This review elaborates on the emerging generation of three-dimensional in vitro models, which comprises spheroids, organoids, and organs-on-chips, which can serve as a stepping stone for advancements in this field.
Collapse
Affiliation(s)
- Joris Van Meenen
- Antwerp Research Group for Ocular Science, Department of Translational Neurosciences, University of Antwerp, Wilrijk, Belgium
| | - Sorcha Ní Dhubhghaill
- Antwerp Research Group for Ocular Science, Department of Translational Neurosciences, University of Antwerp, Wilrijk, Belgium.,Department of Ophthalmology, Antwerp University Hospital, Edegem, Belgium
| | - Bert Van den Bogerd
- Antwerp Research Group for Ocular Science, Department of Translational Neurosciences, University of Antwerp, Wilrijk, Belgium
| | - Carina Koppen
- Antwerp Research Group for Ocular Science, Department of Translational Neurosciences, University of Antwerp, Wilrijk, Belgium.,Department of Ophthalmology, Antwerp University Hospital, Edegem, Belgium
| |
Collapse
|
23
|
Miotti G, Parodi PC, Zeppieri M. Stem cell therapy in ocular pathologies in the past 20 years. World J Stem Cells 2021; 13:366-385. [PMID: 34136071 PMCID: PMC8176844 DOI: 10.4252/wjsc.v13.i5.366] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/12/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023] Open
Abstract
Stem cell therapies are successfully used in various fields of medicine. This new approach of research is also expanding in ophthalmology. Huge investments, resources and important clinical trials have been performed in stem cell research and in potential therapies. In recent years, great strides have been made in genetic research, which permitted and enhanced the differentiation of stem cells. Moreover, the possibility of exploiting stem cells from other districts (such as adipose, dental pulp, bone marrow stem cells, etc.) for the treatment of ophthalmic diseases, renders this topic fascinating. Furthermore, great strides have been made in biomedical engineering, which have proposed new materials and three-dimensional structures useful for cell therapy of the eye. The encouraging results obtained on clinical trials conducted on animals have given a significant boost in the creation of study protocols also in humans. Results are limited to date, but clinical trials continue to evolve. Our attention is centered on the literature reported over the past 20 years, considering animal (the most represented in literature) and human clinical trials, which are limiting. The aim of our review is to present a brief overview of the main types of treatments based on stem cells in the field of ophthalmic pathologies.
Collapse
Affiliation(s)
- Giovanni Miotti
- Department of Plastic Surgery, University Hospital of Udine, Udine 33100, Italy
| | - Pier Camillo Parodi
- Department of Plastic Surgery, University Hospital of Udine, Udine 33100, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, Udine 33100, Italy
| |
Collapse
|
24
|
Lee H, Son MY. Current Challenges Associated with the Use of Human Induced Pluripotent Stem Cell-Derived Organoids in Regenerative Medicine. Int J Stem Cells 2021; 14:9-20. [PMID: 33632980 PMCID: PMC7904522 DOI: 10.15283/ijsc20140] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/23/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
Innovative advances in stem cell research have resulted in the development of organoids, which are widely used as in vitro models of human organ development and for disease. The long-term goals of scientists include the generation of high-quality organoids with properties like those of native organs, and to expand their use to a variety of applications such as drug discovery and organoid-based cell therapy. In particular, the combination of human induced pluripotent stem cell (iPSC)-derived organoids with the recently developed genome engineering, biotechnology serve as an attractive platform in precision medicine. This review briefly summarizes the generation of organoids derived mostly from iPSCs without ethical issues, and describes the applications and technological advances of organoids under their differentiation and culture conditions. We also discuss the approaches to improve the organoid models, and how organoids can recapitulate mature organ systems of the human body for regenerative medicine. Finally, the future perspectives and remaining challenges in the field have been discussed to provide a better understanding of the potential applications of organoids.
Collapse
Affiliation(s)
- Hana Lee
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea.,KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Korea
| | - Mi-Young Son
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea.,KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Korea
| |
Collapse
|
25
|
In vitro reconstructed 3D corneal tissue models for ocular toxicology and ophthalmic drug development. In Vitro Cell Dev Biol Anim 2021; 57:207-237. [PMID: 33544359 DOI: 10.1007/s11626-020-00533-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/18/2020] [Indexed: 12/13/2022]
Abstract
Testing of all manufactured products and their ingredients for eye irritation is a regulatory requirement. In the last two decades, the development of alternatives to the in vivo Draize eye irritation test method has substantially advanced due to the improvements in primary cell isolation, cell culture techniques, and media, which have led to improved in vitro corneal tissue models and test methods. Most in vitro models for ocular toxicology attempt to reproduce the corneal epithelial tissue which consists of 4-5 layers of non-keratinized corneal epithelial cells that form tight junctions, thereby limiting the penetration of chemicals, xenobiotics, and pharmaceuticals. Also, significant efforts have been directed toward the development of more complex three-dimensional (3D) equivalents to study wound healing, drug permeation, and bioavailability. This review focuses on in vitro reconstructed 3D corneal tissue models and their utilization in ocular toxicology as well as their application to pharmacology and ophthalmic research. Current human 3D corneal epithelial cell culture models have replaced in vivo animal eye irritation tests for many applications, and substantial validation efforts are in progress to verify and approve alternative eye irritation tests for widespread use. The validation of drug absorption models and further development of models and test methods for many ophthalmic and ocular disease applications is required.
Collapse
|
26
|
Nomi K, Hayashi R, Ishikawa Y, Kobayashi Y, Katayama T, Quantock AJ, Nishida K. Generation of functional conjunctival epithelium, including goblet cells, from human iPSCs. Cell Rep 2021; 34:108715. [PMID: 33535050 DOI: 10.1016/j.celrep.2021.108715] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/23/2020] [Accepted: 01/12/2021] [Indexed: 12/13/2022] Open
Abstract
The conjunctival epithelium, which covers the sclera (the white of the eye) and lines the inside of the eyelids, is essential for mucin secretion and the establishment of a healthy tear film. Here, we describe human conjunctival development in a self-formed ectodermal autonomous multi-zone (SEAM) of cells that were derived from human-induced pluripotent stem cells (hiPSCs) and mimic whole-eye development. Our data indicate that epidermal growth factor (EGF) drives the generation of cells with a conjunctival epithelial lineage. We also show that individual conjunctival cells can be sorted and reconstituted by cultivation into a functional conjunctival epithelium that includes mucin-producing goblet cells. Keratinocyte growth factor (KGF), moreover, is necessary for the maturation of hiPSC-derived conjunctival epithelium-particularly the goblet cells-indicating key complementary roles of EGF and KGF in directing the differentiation and maturation, respectively, of the human conjunctival epithelium.
Collapse
Affiliation(s)
- Kimihito Nomi
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan; Department of Stem Cells and Applied Medicine, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Ryuhei Hayashi
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan; Department of Stem Cells and Applied Medicine, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan.
| | - Yuki Ishikawa
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan; Department of Stem Cells and Applied Medicine, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yuki Kobayashi
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan; Department of Stem Cells and Applied Medicine, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Tomohiko Katayama
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan; Department of Stem Cells and Applied Medicine, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Andrew J Quantock
- Structural Biophysics Group, School of Optometry and Vision Sciences, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF24 4HQ, Wales, UK
| | - Kohji Nishida
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
27
|
Manafi N, Shokri F, Achberger K, Hirayama M, Mohammadi MH, Noorizadeh F, Hong J, Liebau S, Tsuji T, Quinn PMJ, Mashaghi A. Organoids and organ chips in ophthalmology. Ocul Surf 2020; 19:1-15. [PMID: 33220469 DOI: 10.1016/j.jtos.2020.11.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022]
Abstract
Recent advances have driven the development of stem cell-derived, self-organizing, three-dimensional miniature organs, termed organoids, which mimic different eye tissues including the retina, cornea, and lens. Organoids and engineered microfluidic organ-on-chips (organ chips) are transformative technologies that show promise in simulating the architectural and functional complexity of native organs. Accordingly, they enable exploration of facets of human disease and development not accurately recapitulated by animal models. Together, these technologies will increase our understanding of the basic physiology of different eye structures, enable us to interrogate unknown aspects of ophthalmic disease pathogenesis, and serve as clinically-relevant surrogates for the evaluation of ocular therapeutics. Both the burden and prevalence of monogenic and multifactorial ophthalmic diseases, which can cause visual impairment or blindness, in the human population warrants a paradigm shift towards organoids and organ chips that can provide sensitive, quantitative, and scalable phenotypic assays. In this article, we review the current situation of organoids and organ chips in ophthalmology and discuss how they can be leveraged for translational applications.
Collapse
Affiliation(s)
- Navid Manafi
- Medical Systems Biophysics and Bioengineering, The Leiden Academic Centre for Drug Research (LACDR), Leiden University, 2333CC, Leiden, the Netherlands; Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Fereshteh Shokri
- Department of Epidemiology, Erasmus Medical Center, 3000 CA, Rotterdam, the Netherlands
| | - Kevin Achberger
- Institute of Neuroanatomy & Developmental Biology (INDB), Eberhard Karls University Tübingen, Österbergstrasse 3, 72074, Tübingen, Germany
| | - Masatoshi Hirayama
- Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital, Chiba, 272-8513, Japan; Department of Ophthalmology, School of Medicine, Keio University, Tokyo, 160-8582, Japan
| | - Melika Haji Mohammadi
- Medical Systems Biophysics and Bioengineering, The Leiden Academic Centre for Drug Research (LACDR), Leiden University, 2333CC, Leiden, the Netherlands
| | | | - Jiaxu Hong
- Medical Systems Biophysics and Bioengineering, The Leiden Academic Centre for Drug Research (LACDR), Leiden University, 2333CC, Leiden, the Netherlands; Department of Ophthalmology and Visual Science, Eye, and ENT Hospital, Shanghai Medical College, Fudan University, 83 Fenyang Road, Shanghai, China; Key NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China; Key Laboratory of Myopia, National Health and Family Planning Commission, Shanghai, China
| | - Stefan Liebau
- Institute of Neuroanatomy & Developmental Biology (INDB), Eberhard Karls University Tübingen, Österbergstrasse 3, 72074, Tübingen, Germany
| | - Takashi Tsuji
- Laboratory for Organ Regeneration, RIKEN Center for Biosystems Dynamics Research, Hyogo, 650-0047, Japan; Organ Technologies Inc., Minato, Tokyo, 105-0001, Japan
| | - Peter M J Quinn
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology & Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University. New York, NY, USA; Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center - New York-Presbyterian Hospital, New York, NY, USA.
| | - Alireza Mashaghi
- Medical Systems Biophysics and Bioengineering, The Leiden Academic Centre for Drug Research (LACDR), Leiden University, 2333CC, Leiden, the Netherlands.
| |
Collapse
|
28
|
Expression of a novel brain specific isoform of C3G is regulated during development. Sci Rep 2020; 10:18838. [PMID: 33139841 PMCID: PMC7606606 DOI: 10.1038/s41598-020-75813-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 10/01/2020] [Indexed: 12/13/2022] Open
Abstract
Mice lacking C3G (RapGEF1), a ubiquitously expressed protein essential for neuronal differentiation, show multiple defects in brain development. Function of C3G in neurogenesis is poorly defined. Here, we identify brain specific expression of a novel C3G isoform in mice and humans. This isoform has an insert in the Crk-binding region, generating a polypeptide of 175 kDa, unlike the previously known 140 kDa form expressed in all other tissues. In the adult mouse brain, C3G expression is seen in neurons, but was not detectable in GFAP-positive cells. C3G levels were high in the CA3 region of hippocampus and in mitral cells of olfactory bulb. Neural progenitor cells positive for Doublecortin and Nestin, show expression of C3G. During development, C3G is expressed in precursor cells prior to their differentiation into mature neurons or astrocytes. The 175 kDa as well as 140 kDa forms are seen in embryonic mouse brain, while only the 175 kDa variant is seen in post-natal brain. Human cerebral organoids generated from induced pluripotent stem cells predominantly expressed the 140 kDa polypeptides, and the 175 kDa isoform appeared upon maturation. This study describes developmental regulation and neuronal expression of a brain specific isoform of C3G, a molecule essential for normal development of the mammalian brain.
Collapse
|
29
|
Theerakittayakorn K, Thi Nguyen H, Musika J, Kunkanjanawan H, Imsoonthornruksa S, Somredngan S, Ketudat-Cairns M, Parnpai R. Differentiation Induction of Human Stem Cells for Corneal Epithelial Regeneration. Int J Mol Sci 2020; 21:E7834. [PMID: 33105778 PMCID: PMC7660084 DOI: 10.3390/ijms21217834] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/16/2020] [Accepted: 10/17/2020] [Indexed: 12/13/2022] Open
Abstract
Deficiency of corneal epithelium causes vision impairment or blindness in severe cases. Transplantation of corneal epithelial cells is an effective treatment but the availability of the tissue source for those cells is inadequate. Stem cells can be induced to differentiate to corneal epithelial cells and used in the treatment. Multipotent stem cells (mesenchymal stem cells) and pluripotent stem cells (embryonic stem cells and induced pluripotent stem cells) are promising cells to address the problem. Various protocols have been developed to induce differentiation of the stem cells into corneal epithelial cells. The feasibility and efficacy of both human stem cells and animal stem cells have been investigated for corneal epithelium regeneration. However, some physiological aspects of animal stem cells are different from those of human stem cells, the protocols suited for animal stem cells might not be suitable for human stem cells. Therefore, in this review, only the investigations of corneal epithelial differentiation of human stem cells are taken into account. The available protocols for inducing the differentiation of human stem cells into corneal epithelial cells are gathered and compared. Also, the pathways involving in the differentiation are provided to elucidate the relevant mechanisms.
Collapse
Affiliation(s)
- Kasem Theerakittayakorn
- Embryo Technology and Stem Cell Research Center, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand; (K.T.); (H.T.N.); (J.M.); (S.I.); (S.S.); (M.K.-C.)
| | - Hong Thi Nguyen
- Embryo Technology and Stem Cell Research Center, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand; (K.T.); (H.T.N.); (J.M.); (S.I.); (S.S.); (M.K.-C.)
| | - Jidapa Musika
- Embryo Technology and Stem Cell Research Center, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand; (K.T.); (H.T.N.); (J.M.); (S.I.); (S.S.); (M.K.-C.)
| | - Hataiwan Kunkanjanawan
- Medeze Research and Development Co., Ltd. 28/9 Moo 8, Phutthamonthon Sai 4 Rd., Krathum Lom, Sam Phran, Nakhon Pathom 73220, Thailand;
| | - Sumeth Imsoonthornruksa
- Embryo Technology and Stem Cell Research Center, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand; (K.T.); (H.T.N.); (J.M.); (S.I.); (S.S.); (M.K.-C.)
| | - Sirilak Somredngan
- Embryo Technology and Stem Cell Research Center, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand; (K.T.); (H.T.N.); (J.M.); (S.I.); (S.S.); (M.K.-C.)
| | - Mariena Ketudat-Cairns
- Embryo Technology and Stem Cell Research Center, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand; (K.T.); (H.T.N.); (J.M.); (S.I.); (S.S.); (M.K.-C.)
| | - Rangsun Parnpai
- Embryo Technology and Stem Cell Research Center, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand; (K.T.); (H.T.N.); (J.M.); (S.I.); (S.S.); (M.K.-C.)
| |
Collapse
|
30
|
Ghareeb AE, Lako M, Figueiredo FC. Recent Advances in Stem Cell Therapy for Limbal Stem Cell Deficiency: A Narrative Review. Ophthalmol Ther 2020; 9:809-831. [PMID: 32970311 PMCID: PMC7708613 DOI: 10.1007/s40123-020-00305-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 09/15/2020] [Indexed: 12/13/2022] Open
Abstract
Destruction of the limbus and depletion of limbal stem cells (LSCs), the adult progenitors of the corneal epithelium, leads to limbal stem cell deficiency (LSCD). LSCD is a rare, progressive ocular surface disorder which results in conjunctivalisation and neovascularisation of the corneal surface. Many strategies have been used in the treatment of LSCD, the common goal of which is to regenerate a self-renewing, transparent, and uniform epithelium on the corneal surface. The development of these techniques has frequently resulted from collaboration between stem cell translational scientists and ophthalmologists. Direct transplantation of autologous or allogeneic limbal tissue from a healthy donor eye is regarded by many as the technique of choice. Expansion of harvested LSCs in vitro allows smaller biopsies to be taken from the donor eye and is considered safer and more acceptable to patients. This technique may be utilised in unilateral cases (autologous) or bilateral cases (living related donor). Recently developed, simple limbal epithelial transplant (SLET) can be performed with equally small biopsies but does not require in vitro cell culture facilities. In the case of bilateral LSCD, where autologous limbal tissue is not available, autologous oral mucosa epithelium can be expanded in vitro and transplanted to the diseased eye. Data on long-term outcomes (over 5 years of follow-up) for many of these procedures is needed, and it remains unclear how they produce a self-renewing epithelium without recreating the vital stem cell niche. Bioengineering techniques offer the ability to re-create the physical characteristics of the stem cell niche, while induced pluripotent stem cells offer an unlimited supply of autologous LSCs. In vivo confocal microscopy and anterior segment OCT will complement impression cytology in the diagnosis, staging, and follow-up of LSCD. In this review we analyse recent advances in the pathology, diagnosis, and treatment of LSCD.
Collapse
Affiliation(s)
- Ali E Ghareeb
- Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, UK.,Department of Ophthalmology, Royal Victoria Infirmary, Newcastle Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, UK
| | - Majlinda Lako
- Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | - Francisco C Figueiredo
- Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, UK. .,Department of Ophthalmology, Royal Victoria Infirmary, Newcastle Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, UK.
| |
Collapse
|
31
|
Shiju TM, Carlos de Oliveira R, Wilson SE. 3D in vitro corneal models: A review of current technologies. Exp Eye Res 2020; 200:108213. [PMID: 32890484 DOI: 10.1016/j.exer.2020.108213] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/11/2020] [Accepted: 08/31/2020] [Indexed: 02/07/2023]
Abstract
Three-dimensional (3D) in vitro models are excellent tools for studying complex biological systems because of their physiological similarity to in vivo studies, cost-effectiveness and decreased reliance on animals. The influence of tissue microenvironment on the cells, cell-cell interaction and the cell-matrix interactions can be elucidated in 3D models, which are difficult to mimic in 2D cultures. In order to develop a 3D model, the required cell types are derived from the tissues or stem cells. A 3D tissue/organ model typically includes all the relevant cell types and the microenvironment corresponding to that tissue/organ. For instance, a full corneal 3D model is expected to have epithelial, stromal, endothelial and nerve cells, along with the extracellular matrix and membrane components associated with the cells. Although it is challenging to develop a corneal 3D model, several attempts have been made and various technologies established which closely mimic the in vivo environment. In this review, three major technologies are highlighted: organotypic cultures, organoids and 3D bioprinting. Also, several combinations of organotypic cultures, such as the epithelium and stroma or endothelium and neural cultures are discussed, along with the disease relevance and potential applications of these models. In the future, new biomaterials will likely promote better cell-cell and cell-matrix interactions in organotypic corneal cultures.
Collapse
|
32
|
Bansal M, Tandon R, Saxena R, Sharma A, Sen S, Kishore A, Venkatesh P, Maiti S, Chakraborty D. Ophthalmic genetics practice and research in India: Vision in 2020. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2020; 184:718-727. [PMID: 32865332 DOI: 10.1002/ajmg.c.31827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/19/2020] [Accepted: 07/28/2020] [Indexed: 12/13/2022]
Abstract
Ophthalmic genetics is a much needed and growing area in India. Ethnic diversity, with a high degree of consanguinity, has led to a high prevalence of genetic disorders in the country. As the second most populous country in the world, this naturally results in a significant number of affected people overall. Practice involves coherent association between ophthalmologists, genetic counselor and pediatricians. Eye genetics in India in recent times has witnessed advanced research using cutting edge diagnostics, next generation sequencing (NGS) approaches, stem cell therapies, gene therapy and genomic editing. This article will highlight the studies reporting genetic variations in the country, challenges in practice, and the latest advances in ophthalmic genetic research in India.
Collapse
Affiliation(s)
- Mayank Bansal
- Council for Scientific and Industrial Research (CSIR), Institute of Genomics and Integrative Biology (IGIB), New Delhi, India.,Academy of Scientific and Innovative Research (AcSIR), Delhi, India.,Department of Ophthalmology, Fortis Memorial Research Institute, Gurugram, India
| | - Radhika Tandon
- Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Rohit Saxena
- Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Arundhati Sharma
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Sagnik Sen
- Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Alisha Kishore
- Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Pradeep Venkatesh
- Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Souvik Maiti
- Council for Scientific and Industrial Research (CSIR), Institute of Genomics and Integrative Biology (IGIB), New Delhi, India.,Academy of Scientific and Innovative Research (AcSIR), Delhi, India
| | - Debojyoti Chakraborty
- Council for Scientific and Industrial Research (CSIR), Institute of Genomics and Integrative Biology (IGIB), New Delhi, India.,Academy of Scientific and Innovative Research (AcSIR), Delhi, India
| |
Collapse
|
33
|
Bioengineering of Human Corneal Endothelial Cells from Single- to Four-Dimensional Cultures. CURRENT OPHTHALMOLOGY REPORTS 2020. [DOI: 10.1007/s40135-020-00244-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
34
|
Bai J, Wang C. Organoids and Microphysiological Systems: New Tools for Ophthalmic Drug Discovery. Front Pharmacol 2020; 11:407. [PMID: 32317971 PMCID: PMC7147294 DOI: 10.3389/fphar.2020.00407] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 03/18/2020] [Indexed: 12/13/2022] Open
Abstract
Organoids are adept at preserving the inherent complexity of a given cellular environment and when integrated with engineered micro-physiological systems (MPS) present distinct advantages for simulating a precisely controlled geometrical, physical, and biochemical micro-environment. This then allows for real-time monitoring of cell-cell interactions. As a result, the two aforementioned technologies hold significant promise and potential in studying ocular physiology and diseases by replicating specific eye tissue microstructures in vitro. This miniaturized review begins with defining the science behind organoids/MPS and subsequently introducing methods for generating organoids and engineering MPS. Furthermore, we will discuss the current state of organoids and MPS models in retina, cornea surrogates, and other ocular tissue, in regards to physiological/disease conditions. Finally, future prospective on organoid/MPS will be covered here. Organoids and MPS technologies closely recapture the in vivo microenvironment and thusly will continue to provide new understandings in organ functions and novel approaches to drug development.
Collapse
Affiliation(s)
- Jing Bai
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau
| |
Collapse
|
35
|
Sharma A, Sances S, Workman MJ, Svendsen CN. Multi-lineage Human iPSC-Derived Platforms for Disease Modeling and Drug Discovery. Cell Stem Cell 2020; 26:309-329. [PMID: 32142662 PMCID: PMC7159985 DOI: 10.1016/j.stem.2020.02.011] [Citation(s) in RCA: 173] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human induced pluripotent stem cells (hiPSCs) provide a powerful platform for disease modeling and have unlocked new possibilities for understanding the mechanisms governing human biology, physiology, and genetics. However, hiPSC-derivatives have traditionally been utilized in two-dimensional monocultures, in contrast to the multi-systemic interactions that influence cells in the body. We will discuss recent advances in generating more complex hiPSC-based systems using three-dimensional organoids, tissue-engineering, microfluidic organ-chips, and humanized animal systems. While hiPSC differentiation still requires optimization, these next-generation multi-lineage technologies can augment the biomedical researcher's toolkit and enable more realistic models of human tissue function.
Collapse
Affiliation(s)
- Arun Sharma
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Samuel Sances
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Michael J Workman
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
36
|
Heidari M, Noorizadeh F, Wu K, Inomata T, Mashaghi A. Dry Eye Disease: Emerging Approaches to Disease Analysis and Therapy. J Clin Med 2019; 8:jcm8091439. [PMID: 31514344 PMCID: PMC6780511 DOI: 10.3390/jcm8091439] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/01/2019] [Accepted: 09/06/2019] [Indexed: 12/11/2022] Open
Abstract
Dry eye disease (DED) is among the most common ocular disorders affecting tens of millions of individuals worldwide; however, the condition remains incompletely understood and treated. Valuable insights have emerged from multidisciplinary approaches, including immunometabolic analyses, microbiome analyses, and bioengineering. Furthermore, we have seen new developments in clinical assessment approaches and treatment strategies in the recent past. Here, we review the emerging frontiers in the pathobiology and clinical management of DED.
Collapse
Affiliation(s)
- Mostafa Heidari
- Basir Eye Health Research Center, Tehran 1418643561, Iran.
- Farabi Eye Hospital, Department of Ophthalmology and Eye Research Center, Tehran University of Medical Sciences, Tehran 133661635, Iran.
| | | | - Kevin Wu
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, Ophthalmic Consultation Service, New York, NY 10029, USA
- New York Eye and Ear Infirmary of Mount Sinai, New York, NY 10003, USA
| | - Takenori Inomata
- Department of Ophthalmology, Juntendo University Faculty of Medicine, Tokyo 1130033, Japan.
- Department of Strategic Operating Room Management and Improvement, Juntendo University Faculty of Medicine, Tokyo 1130033, Japan.
| | - Alireza Mashaghi
- Systems Biomedicine and Pharmacology Division, Leiden Academic Centre for Drug Research, Leiden University, 2333CC Leiden, The Netherlands.
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA.
- Department of Ophthalmology, Shanghai Medical College, Fudan University, Shanghai 200000, China.
| |
Collapse
|
37
|
Wright CB, Becker SM, Low LA, Tagle DA, Sieving PA. Improved Ocular Tissue Models and Eye-On-A-Chip Technologies Will Facilitate Ophthalmic Drug Development. J Ocul Pharmacol Ther 2019; 36:25-29. [PMID: 31166829 PMCID: PMC6985761 DOI: 10.1089/jop.2018.0139] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 03/20/2019] [Indexed: 12/13/2022] Open
Abstract
In this study, we describe efforts by the National Eye Institute (NEI) and National Center for Advancing Translational Science (NCATS) to catalyze advances in 3-dimensional (3-D) ocular organoid and microphysiological systems (MPS). We reviewed the recent literature regarding ocular organoids and tissue chips. Animal models, 2-dimensional cell culture models, and postmortem human tissue samples provide the vision research community with insights critical to understanding pathophysiology and therapeutic development. The advent of induced pluripotent stem cell technologies provide researchers with enticing new approaches and tools that augment study in more traditional models to provide the scientific community with insights that have previously been impossible to obtain. Efforts by the National Institutes of Health (NIH) have already accelerated the pace of scientific discovery, and recent advances in ocular organoid and MPS modeling approaches have opened new avenues of investigation. In addition to more closely recapitulating the morphologies and physiological responses of in vivo human tissue, key breakthroughs have been made in the past year to resolve long-standing scientific questions regarding tissue development, molecular signaling, and pathophysiological mechanisms that promise to provide advances critical to therapeutic development and patient care. 3-D tissue culture modeling and MPS offer platforms for future high-throughput testing of therapeutic candidates and studies of gene interactions to improve models of complex genetic diseases with no well-defined etiology, such as age-related macular degeneration and Fuchs' dystrophy.
Collapse
Affiliation(s)
- Charles B. Wright
- National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Steven M. Becker
- National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Lucie A. Low
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland
| | - Danilo A. Tagle
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland
| | - Paul A. Sieving
- National Eye Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
38
|
Xia X, Li F, He J, Aji R, Gao D. Organoid technology in cancer precision medicine. Cancer Lett 2019; 457:20-27. [PMID: 31078736 DOI: 10.1016/j.canlet.2019.04.039] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/22/2019] [Accepted: 04/25/2019] [Indexed: 12/13/2022]
Abstract
Organoid technology has been remarkably improved over the last decade. Various organoids have been derived from different types of tissues and recapitulate their organ-specific gene expression signatures, particular tissue spatial structures and functions of their original tissue. The patient-derived organoids (PDOs) have been used to elucidate crucial scientific questions, including the relationships between genetic/epigenetic alterations and drug responses, cell plasticity during disease progressions, and mechanisms of drug resistances. With the great expectations, PDOs will be widely used to facilitate the personalized medical decisions, which have the potential to profoundly improve patient outcomes. In this review, we will discuss the developmental details, current achievements, applications and challenges of organoid technology in precision cancer medicine.
Collapse
Affiliation(s)
- Xinyi Xia
- State Key Laboratory of Cell Biology, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China; CAS Key Laboratory of Systems Biology, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Fei Li
- State Key Laboratory of Cell Biology, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China; CAS Key Laboratory of Systems Biology, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Juan He
- State Key Laboratory of Cell Biology, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China; CAS Key Laboratory of Systems Biology, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Rebiguli Aji
- State Key Laboratory of Cell Biology, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China; CAS Key Laboratory of Systems Biology, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Dong Gao
- State Key Laboratory of Cell Biology, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China; CAS Key Laboratory of Systems Biology, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.
| |
Collapse
|
39
|
Prospects and modalities for the treatment of genetic ocular anomalies. Hum Genet 2019; 138:1019-1026. [DOI: 10.1007/s00439-018-01968-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 12/24/2018] [Indexed: 12/13/2022]
|
40
|
Haderspeck JC, Chuchuy J, Kustermann S, Liebau S, Loskill P. Organ-on-a-chip technologies that can transform ophthalmic drug discovery and disease modeling. Expert Opin Drug Discov 2018; 14:47-57. [DOI: 10.1080/17460441.2019.1551873] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Jasmin C. Haderspeck
- Institute of Neuroanatomy & Developmental Biology (INDB), Eberhard Karls University Tübingen, Tübingen, Germany
| | - Johanna Chuchuy
- Department of Women’s Health, Research Institute for Women’s Health, Eberhard Karls University Tübingen, Tübingen, Germany
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
| | - Stefan Kustermann
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Stefan Liebau
- Institute of Neuroanatomy & Developmental Biology (INDB), Eberhard Karls University Tübingen, Tübingen, Germany
| | - Peter Loskill
- Department of Women’s Health, Research Institute for Women’s Health, Eberhard Karls University Tübingen, Tübingen, Germany
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
| |
Collapse
|
41
|
Abstract
Human-induced pluripotent stem cells (hiPSCs) provide a personalized approach to study conditions and diseases including those of the eye that lack appropriate animal models to facilitate the development of novel therapeutics. Corneal disease is one of the most common causes of blindness. Hence, significant efforts are made to develop novel therapeutic approaches including stem cell-derived strategies to replace the diseased or damaged corneal tissues, thus restoring the vision. The use of adult limbal stem cells in the management of corneal conditions has been clinically successful. However, its limited availability and phenotypic plasticity necessitate the need for alternative stem cell sources to manage corneal conditions. Mesenchymal and embryonic stem cell-based approaches are being explored; nevertheless, their limited differentiation potential and ethical concerns have posed a significant hurdle in its clinical use. hiPSCs have emerged to fill these technical and ethical gaps to render clinical utility. In this review, we discuss and summarize protocols that have been devised so far to direct differentiation of human pluripotent stem cells (hPSCs) to different corneal cell phenotypes. With the summarization, our review intends to facilitate an understanding which would allow developing efficient and robust protocols to obtain specific corneal cell phenotype from hPSCs for corneal disease modeling and for the clinics to treat corneal diseases and injury.
Collapse
Affiliation(s)
| | - Rohit Shetty
- Cornea and Refractive Surgery, Narayana Nethralaya, Bengaluru, India
| | - Arkasubhra Ghosh
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bengaluru, India
| |
Collapse
|
42
|
Hongisto H, Vattulainen M, Ilmarinen T, Mikhailova A, Skottman H. Efficient and Scalable Directed Differentiation of Clinically Compatible Corneal Limbal Epithelial Stem Cells from Human Pluripotent Stem Cells. J Vis Exp 2018. [PMID: 30417867 DOI: 10.3791/58279] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Corneal limbal epithelial stem cells (LESCs) are responsible for continuously renewing the corneal epithelium, and thus maintaining corneal homeostasis and visual clarity. Human pluripotent stem cell (hPSC)-derived LESCs provide a promising cell source for corneal cell replacement therapy. Undefined, xenogeneic culture and differentiation conditions cause variation in research results and impede the clinical translation of hPSC-derived therapeutics. This protocol provides a reproducible and efficient method for hPSC-LESC differentiation under xeno- and feeder cell-free conditions. Firstly, monolayer culture of undifferentiated hPSC on recombinant laminin-521 (LN-521) and defined hPSC medium serves as a foundation for robust production of high-quality starting material for differentiations. Secondly, a rapid and simple hPSC-LESC differentiation method yields LESC populations in only 24 days. This method includes a four-day surface ectodermal induction in suspension with small molecules, followed by adherent culture phase on LN-521/collagen IV combination matrix in defined corneal epithelial differentiation medium. Cryostoring and extended differentiation further purifies the cell population and enables banking of the cells in large quantities for cell therapy products. The resulting high-quality hPSC-LESCs provide a potential novel treatment strategy for corneal surface reconstruction to treat limbal stem cell deficiency (LSCD).
Collapse
Affiliation(s)
- Heidi Hongisto
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere;
| | - Meri Vattulainen
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere
| | - Tanja Ilmarinen
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere
| | - Alexandra Mikhailova
- Department of Ophthalmology, SILK, Faculty of Medicine and Life Sciences, University of Tampere
| | - Heli Skottman
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere
| |
Collapse
|
43
|
Yang J, Park JW, Zheng D, Xu RH. Universal Corneal Epithelial-Like Cells Derived from Human Embryonic Stem Cells for Cellularization of a Corneal Scaffold. Transl Vis Sci Technol 2018; 7:23. [PMID: 30323996 PMCID: PMC6181193 DOI: 10.1167/tvst.7.5.23] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/07/2018] [Indexed: 12/13/2022] Open
Abstract
Purpose We generated universal corneal epithelial cells (CEC) from human embryonic stem cells (hESC) by genetically removing human leukocyte antigens (HLA) class I from the cell surface. Methods The serum-free, growth factor-free, and defined medium E6 was used to differentiate hESC to CEC. Decellularized murine corneas were recellularized with hESC-derived CEC. Using CRISPR/Cas9, β-2-microglobulin (B2M) was deleted in hESC to block the assembly of HLA class-I antigens on the cell surface to generate B2M−/− CEC. Results E6 alone was sufficient to allow hESC differentiation to CEC. A time-course analysis of the global gene expression of the differentiating cells indicates that the differentiation closely resembles the corneal development in vivo. The hESC-CEC were highly proliferative, and could form multilayer epithelium in decellularized murine cornea, retain its transparency, and form intact tight junctions on its surface. As reported before, B2M knockout led to the absence of HLA class-I on the cell surface of hESC and subsequently derived CEC following stimulation with inflammatory factors. Moreover, B2M−/− CEC, following transplantation into mouse eyes, caused less T-cell infiltration in the limbal region of the eye than the wild-type control. Conclusions CEC can be derived from hESC via a novel and simple protocol free of any proteins, hESC-CEC seeded on decellularized animal cornea form tight junctions and allow light transmittance, and B2M−/− CEC are hypoimmunogenic both in vitro and in vivo. Translational Relevance B2M−/− hESC-CEC can be an unlimited and universal therapy for corneal repair in patients of any HLA type.
Collapse
Affiliation(s)
- Juan Yang
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Jung Woo Park
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Dejin Zheng
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Ren-He Xu
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
44
|
Yokokura S, Tanaka Y. Recent Advances in Biosynthetic Corneal Substitutes. CURRENT OPHTHALMOLOGY REPORTS 2018. [DOI: 10.1007/s40135-018-0180-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
45
|
Hongisto H, Ilmarinen T, Vattulainen M, Mikhailova A, Skottman H. Xeno- and feeder-free differentiation of human pluripotent stem cells to two distinct ocular epithelial cell types using simple modifications of one method. Stem Cell Res Ther 2017; 8:291. [PMID: 29284513 PMCID: PMC5747074 DOI: 10.1186/s13287-017-0738-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/13/2017] [Accepted: 11/28/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Human pluripotent stem cells (hPSCs) provide a promising cell source for ocular cell replacement therapy, but often lack standardized and xenogeneic-free culture and differentiation protocols. We aimed to develop a xeno- and feeder cell-free culture system for undifferentiated hPSCs along with efficient methods to derive ocular therapy target cells: retinal pigment epithelial (RPE) cells and corneal limbal epithelial stem cells (LESCs). METHODS Multiple genetically distinct hPSC lines were adapted to a defined, xeno-, and feeder-free culture system of Essential 8™ medium and laminin-521 matrix. Thereafter, two-stage differentiation methods toward ocular epithelial cells were established utilizing xeno-free media and a combination of extracellular matrix proteins. Both differentiation methods shared the same basal elements, using only minor inductive modifications during early differentiation towards desired cell lineages. The resulting RPE cells and LESCs were characterized after several independent differentiation experiments and recovery after xeno-free cryopreservation. RESULTS The defined, xeno-, and feeder-free culture system provided a robust means to generate high-quality hPSCs with chromosomal stability limited to early passages. Inductive cues introduced during the first week of differentiation had a substantial effect on lineage specification, cell survival, and even mature RPE properties. Derivative RPE formed functional epithelial monolayers with mature tight junctions and expression of RPE genes and proteins, as well as phagocytosis and key growth factor secretion capacity after 9 weeks of maturation on inserts. Efficient LESC differentiation led to cell populations expressing LESC markers such as p40/p63α by day 24. Finally, we established xeno-free cryobanking protocols for pluripotent hPSCs, hPSC-RPE cells, and hPSC-LESCs, and demonstrated successful recovery after thawing. CONCLUSIONS We propose methods for efficient and scalable, directed differentiation of high-quality RPE cells and LESCs. The two clinically relevant cell types are generated with simple inductive modification of the same basal method, followed by adherent culture, passaging, and cryobanking.
Collapse
Affiliation(s)
- Heidi Hongisto
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere, Arvo Ylpön katu 34, 33520, Tampere, Finland.
| | - Tanja Ilmarinen
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere, Arvo Ylpön katu 34, 33520, Tampere, Finland
| | - Meri Vattulainen
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere, Arvo Ylpön katu 34, 33520, Tampere, Finland
| | - Alexandra Mikhailova
- Department of Ophthalmology, SILK, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland.,Finnish Federation of the Visually Impaired, Helsinki, Finland
| | - Heli Skottman
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere, Arvo Ylpön katu 34, 33520, Tampere, Finland
| |
Collapse
|