1
|
Zhao L, Shi Z, Qi X, Wang J, Yu M, Dong M, Wang F, Zhou Q, Wang T, Shi W. Corneal stromal structure replicating humanized hydrogel patch for sutureless repair of deep anterior-corneal defect. Biomaterials 2025; 313:122754. [PMID: 39197237 DOI: 10.1016/j.biomaterials.2024.122754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/04/2024] [Accepted: 08/11/2024] [Indexed: 09/01/2024]
Abstract
A critical shortage of donor corneas exists worldwide. Hydrogel patches with a biological architecture and functions that simulate those of native corneas have garnered considerable attention. This study introduces a stromal structure replicating corneal patch (SRCP) composed of a decellularized cornea-templated nanotubular skeleton, recombinant human collagen, and methacrylated gelatin, exhibiting a similar ultrastructure and transmittance (above 80 %) to natural cornea. The SRCP is superior to the conventional recombinant human collagen patch in terms of biomechanical properties and resistance to enzymatic degradation. Additionally, SRCP promotes corneal epithelial and stromal cell migration while preventing the trans-differentiation of stromal cells into myofibroblasts. When applied to an ocular surface (37 °C), SRCP releases methacrylated gelatin, which robustly binds SRCP to the corneal stroma after activation by 405 nm light. Compared to gelatin-based photocurable hydrogel, the SRCP better supports the restoration of normal corneal curvature and withstands deformation under an elevated intraocular pressure (100 mmHg). In an in vivo deep anterior-corneal defect model, SRCP facilitated epithelial healing and vision recovery within 2 weeks, maintained graft structural stability, and inhibited stromal scarring at 4 weeks post-operation. The ideal performance of the SRCP makes it a promising humanized corneal equivalent for sutureless clinical applications.
Collapse
Affiliation(s)
- Long Zhao
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China
| | - Zhen Shi
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China
| | - Xia Qi
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China
| | - Jingting Wang
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China
| | - Mengmeng Yu
- Eye Institute of Shandong First Medical University, Eye Hospital of Shandong First Medical University (Shandong Eye Hospital), Jinan, 250021, China
| | - Muchen Dong
- Eye Institute of Shandong First Medical University, Eye Hospital of Shandong First Medical University (Shandong Eye Hospital), Jinan, 250021, China
| | - Fuyan Wang
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China
| | - Qingjun Zhou
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China
| | - Ting Wang
- Eye Institute of Shandong First Medical University, Eye Hospital of Shandong First Medical University (Shandong Eye Hospital), Jinan, 250021, China.
| | - Weiyun Shi
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071, China; Eye Institute of Shandong First Medical University, Eye Hospital of Shandong First Medical University (Shandong Eye Hospital), Jinan, 250021, China.
| |
Collapse
|
2
|
Rafiei S, Ghanbari-Abdolmaleki M, Zeinali R, Heidari-Keshel S, Rahimi A, Royanian F, Zaeifi D, Taheri K, Pourtaghi K, Khaleghi M, Biazar E. Silk fibroin/vitreous humor hydrogel scaffold modified by a carbodiimide crosslinker for wound healing. Biopolymers 2024; 115:e23612. [PMID: 38994706 DOI: 10.1002/bip.23612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/13/2024]
Abstract
Natural-derived biomaterials can be used as substrates for the growth, proliferation, and differentiation of cells. In this study, bovine vitreous humor as a biological material was cross-linked to silk fibroin with different concentration ratios to design a suitable substrate for corneal tissue regeneration. The cross-linked samples were evaluated with different analyses such as structural, physical (optical, swelling, and degradation), mechanical, and biological (viability, cell adhesion) assays. The results showed that all samples had excellent transparency, especially those with higher silk fibroin content. Increasing the ratio of vitreous humor to silk fibroin decreased mechanical strength and increased swelling and degradation, respectively. There was no significant difference in the toxicity of the samples, and with the increase in vitreous humor ratio, adhesion and cell proliferation increased. Generally, silk fibroin with vitreous humor can provide desirable characteristics as a transparent film for corneal wound healing.
Collapse
Affiliation(s)
- Sepideh Rafiei
- Tissue Engineering Group, Department of Biomedical Engineering, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | | | - Reza Zeinali
- Group of Molecular and Industrial Biotechnology, Department of Chemical Engineering, Universität Politècnica de Catalunya, Terrassa, Spain
| | - Saeed Heidari-Keshel
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azam Rahimi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farima Royanian
- Tissue Engineering Group, Department of Biomedical Engineering, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Davood Zaeifi
- Department of Cellular and Molecular Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Kiana Taheri
- Tissue Engineering Group, Department of Biomedical Engineering, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Kimia Pourtaghi
- Tissue Engineering Group, Department of Biomedical Engineering, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Maryam Khaleghi
- Tissue Engineering Group, Department of Biomedical Engineering, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Esmaeil Biazar
- Tissue Engineering Group, Department of Biomedical Engineering, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| |
Collapse
|
3
|
Kantawala B, Shariff S, Ramadan N, Fawaz V, Hassan Y, Mugisha N, Yenkoyan K, Nazir A, Uwishema O. Revolutionizing neurotherapeutics: blood-brain barrier-on-a-chip technologies for precise drug delivery. Ann Med Surg (Lond) 2024; 86:2794-2804. [PMID: 38694300 PMCID: PMC11060226 DOI: 10.1097/ms9.0000000000001887] [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: 11/23/2023] [Accepted: 02/23/2024] [Indexed: 05/04/2024] Open
Abstract
Introduction The blood-brain barrier (BBB) is a critical neurovascular unit regulating substances' passage from the bloodstream to the brain. Its selective permeability poses significant challenges in drug delivery for neurological disorders. Conventional methods often fail due to the BBB's complex structure. Aim The study aims to shed light on their pivotal role in revolutionizing neurotherapeutics and explores the transformative potential of BBB-on-a-Chip technologies in drug delivery research to comprehensively review BBB-on-a-chip technologies, focusing on their design, and substantiate advantages over traditional models. Methods A detailed analysis of existing literature and experimental data pertaining to BBB-on-a-Chip technologies was conducted. Various models, their physiological relevance, and innovative design considerations were examined through databases like Scopus, EbscoHost, PubMed Central, and Medline. Case studies demonstrating enhanced drug transport through BBB-on-a-Chip models were also reviewed, highlighting their potential impact on neurological disorders. Results BBB-on-a-Chip models offer a revolutionary approach, accurately replicating BBB properties. These microphysiological systems enable high-throughput screening, real-time monitoring of drug transport, and precise localization of drugs. Case studies demonstrate their efficacy in enhancing drug penetration, offering potential therapies for diseases like Parkinson's and Alzheimer's. Conclusion BBB-on-a-Chip models represent a transformative milestone in drug delivery research. Their ability to replicate BBB complexities, offer real-time monitoring, and enhance drug transport holds immense promise for neurological disorders. Continuous research and development are imperative to unlock BBB-on-a-Chip models' full potential, ushering in a new era of targeted, efficient, and safer drug therapies for challenging neurological conditions.
Collapse
Affiliation(s)
- Burhan Kantawala
- Oli Health Magazine Organization, Research and Education
- Neuroscience Laboratory, Cobrain Centre
| | - Sanobar Shariff
- Oli Health Magazine Organization, Research and Education
- Neuroscience Laboratory, Cobrain Centre
| | - Nagham Ramadan
- Oli Health Magazine Organization, Research and Education
- Faculty of Medicine
| | - Violette Fawaz
- Oli Health Magazine Organization, Research and Education
- Faculty of Pharmacy, Beirut Arab University, Beirut, Lebanon
| | - Youmna Hassan
- Oli Health Magazine Organization, Research and Education
- Faculty of Medicine and Surgery, Ahfad University for Women, Omdurman, Sudan
| | - Nadine Mugisha
- Oli Health Magazine Organization, Research and Education
- Faculty of Global Surgery, University of Global Health Equity, Kigali, Rwanda
| | - Konstantin Yenkoyan
- Neuroscience Laboratory, Cobrain Centre
- Department of Biochemistry, Yerevan State Medical University named after Mkhitar Heratsi, Yerevan, Armenia
| | - Abubakar Nazir
- Oli Health Magazine Organization, Research and Education
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | | |
Collapse
|
4
|
Dos Santos FV, Siqueira RL, de Morais Ramos L, Yoshioka SA, Branciforti MC, Correa DS. Silk fibroin-derived electrospun materials for biomedical applications: A review. Int J Biol Macromol 2024; 254:127641. [PMID: 37913875 DOI: 10.1016/j.ijbiomac.2023.127641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/14/2023] [Accepted: 10/22/2023] [Indexed: 11/03/2023]
Abstract
Electrospinning is a versatile technique for fabricating polymeric fibers with diameters ranging from micro- to nanoscale, exhibiting multiple morphologies and arrangements. By combining silk fibroin (SF) with synthetic and/or natural polymers, electrospun materials with outstanding biological, chemical, electrical, physical, mechanical, and optical properties can be achieved, fulfilling the evolving biomedical demands. This review highlights the remarkable versatility of SF-derived electrospun materials, specifically focusing on their application in tissue regeneration (including cartilage, cornea, nerves, blood vessels, bones, and skin), disease treatment (such as cancer and diabetes), and the development of controlled drug delivery systems. Additionally, we explore the potential future trends in utilizing these nanofibrous materials for creating intelligent biomaterials, incorporating biosensors and wearable sensors for monitoring human health, and also discuss the bottlenecks for its widespread use. This comprehensive overview illuminates the significant impact and exciting prospects of SF-derived electrospun materials in advancing biomedical research and applications.
Collapse
Affiliation(s)
- Francisco Vieira Dos Santos
- Nanotechnology National Laboratory for Agriculture, Embrapa Instrumentação, 13560-970 São Carlos, SP, Brazil; Materials Engineering Department, São Carlos School of Engineering, University of São Paulo, 13563-120 São Carlos, SP, Brazil
| | - Renato Luiz Siqueira
- Materials Engineering Department, Federal University of São Carlos, 13565-905 São Carlos, SP, Brazil
| | - Lucas de Morais Ramos
- São Carlos Institute of Physics, University of São Paulo, 13560-970 São Carlos, SP, Brazil
| | - Sérgio Akinobu Yoshioka
- Laboratory of Biochemistry and Biomaterials, São Carlos Institute of Chemistry, University of São Paulo, 13560-970 São Carlos, SP, Brazil
| | - Márcia Cristina Branciforti
- Materials Engineering Department, São Carlos School of Engineering, University of São Paulo, 13563-120 São Carlos, SP, Brazil
| | - Daniel Souza Correa
- Nanotechnology National Laboratory for Agriculture, Embrapa Instrumentação, 13560-970 São Carlos, SP, Brazil; Materials Engineering Department, São Carlos School of Engineering, University of São Paulo, 13563-120 São Carlos, SP, Brazil.
| |
Collapse
|
5
|
Bhattacharjee P, Madden PW, Patriarca E, Ahearne M. Optimization and evaluation of oxygen-plasma-modified, aligned, poly (Є-caprolactone) and silk fibroin nanofibrous scaffold for corneal stromal regeneration. BIOMATERIALS AND BIOSYSTEMS 2023; 12:100083. [PMID: 37731910 PMCID: PMC10507194 DOI: 10.1016/j.bbiosy.2023.100083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/28/2023] [Accepted: 09/02/2023] [Indexed: 09/22/2023] Open
Abstract
The shortage of human donor corneas for transplantation necessitates the exploration of tissue engineering approaches to develop corneal substitutes. However, these substitutes must possess the necessary strength, transparency, and ability to regulate cell behaviour before they can be used in patients. In this study, we investigated the effectiveness of an oxygen plasma surface-modified poly-ε-caprolactone (PCL) combined with silk fibroin (SF) nanofibrous scaffold for corneal stromal regeneration. To fabricate the electrospun scaffolds, PCL and SF blends were used on a rotating mandrel. The optimization of the blend aimed to replicate the structural and functional properties of the human cornea, focusing on nanofibre alignment, mechanical characteristics, and in vitro cytocompatibility with human corneal stromal keratocytes. Surface modification of the scaffold resulted in improved transparency and enhanced cell interaction. Based on the evaluation, a composite nanofibrous scaffold with a 1:1 blend of PCL and SF was selected for a more comprehensive analysis. The biological response of keratocytes to the scaffold was assessed through cellular adhesion, proliferation, cytoskeletal organization, gene expression, and immunocytochemical staining. The scaffold facilitated the adhesion of corneal stromal cells, supporting cell proliferation, maintaining normal cytoskeletal organization, and promoting increased expression of genes associated with healthy corneal stromal keratocytes. These findings highlight the potential of a surface-modified PCL/SF blend (1:1) as a promising scaffolding material for corneal stromal regeneration. The developed scaffold not only demonstrated favourable biological interactions with corneal stromal cells but also exhibited characteristics aligned with the requirements for successful corneal tissue engineering. Further research and refinement of these constructs could lead to significant advancements in addressing the shortage of corneas for transplantation, ultimately improving the treatment outcomes for patients in need.
Collapse
Affiliation(s)
- Promita Bhattacharjee
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, University of Dublin, Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, University of Dublin, Dublin, Ireland
| | - Peter W. Madden
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, University of Dublin, Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, University of Dublin, Dublin, Ireland
| | - Enzo Patriarca
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, University of Dublin, Dublin, Ireland
| | - Mark Ahearne
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, University of Dublin, Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, University of Dublin, Dublin, Ireland
| |
Collapse
|
6
|
Al-Atawi S. Three-dimensional bioprinting in ophthalmic care. Int J Ophthalmol 2023; 16:1702-1711. [PMID: 37854366 PMCID: PMC10559024 DOI: 10.18240/ijo.2023.10.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 05/04/2023] [Indexed: 10/20/2023] Open
Abstract
Three-dimensional (3D) bioprinting is widely used in ophthalmic clinic, including in diagnosis, surgery, prosthetics, medications, drug development and delivery, and medical education. Articles published in 2011-2022 into bioinks, printing technologies, and bioprinting applications in ophthalmology were reviewed and the strengths and limitations of bioprinting in ophthalmology highlighted. The review highlighted the trade-offs of printing technologies and bioinks in respect to, among others, material type cost, throughput, gelation technique, cell density, cell viability, resolution, and printing speed. There is already widespread ophthalmological application of bioprinting outside clinical settings, including in educational modelling, retinal imaging/visualization techniques and drug design/testing. In clinical settings, bioprinting has already found application in pre-operatory planning. Even so, the findings showed that even with its immense promise, actual translation to clinical applications remains distant, but relatively closer for the corneal (except stromal) tissues, epithelium, endothelium, and conjunctiva, than it was for the retina. This review similarly reflected on the critical on the technical, practical, ethical, and cost barrier to rapid progress of bioprinting in ophthalmology, including accessibility to the most sophisticated bioprinting technologies, choice, and suitability of bioinks, tissue viability and storage conditions. The extant research is encouraging, but more work is clearly required for the push towards clinical translation of research.
Collapse
Affiliation(s)
- Saleha Al-Atawi
- Al-baha University, Applied Medical Science, Al-Aqiaq, AlBaha 4781, Saudi Arabia
| |
Collapse
|
7
|
Sahoo JK, Hasturk O, Falcucci T, Kaplan DL. Silk chemistry and biomedical material designs. Nat Rev Chem 2023; 7:302-318. [PMID: 37165164 DOI: 10.1038/s41570-023-00486-x] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2023] [Indexed: 05/12/2023]
Abstract
Silk fibroin has applications in different medical fields such as tissue engineering, regenerative medicine, drug delivery and medical devices. Advances in silk chemistry and biomaterial designs have yielded exciting tools for generating new silk-based materials and technologies. Selective chemistries can enhance or tune the features of silk, such as mechanics, biodegradability, processability and biological interactions, to address challenges in medically relevant materials (hydrogels, films, sponges and fibres). This Review details the design and utility of silk biomaterials for different applications, with particular focus on chemistry. This Review consists of three segments: silk protein fundamentals, silk chemistries and functionalization mechanisms. This is followed by a description of different crosslinking chemistries facilitating network formation, including the formation of composite biomaterials. Utility in the fields of tissue engineering, drug delivery, 3D printing, cell coatings, microfluidics and biosensors are highlighted. Looking to the future, we discuss silk biomaterial design strategies to continue to improve medical outcomes.
Collapse
Affiliation(s)
| | - Onur Hasturk
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Thomas Falcucci
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA.
| |
Collapse
|
8
|
Kumar Sahi A, Gundu S, Kumari P, Klepka T, Sionkowska A. Silk-Based Biomaterials for Designing Bioinspired Microarchitecture for Various Biomedical Applications. Biomimetics (Basel) 2023; 8:biomimetics8010055. [PMID: 36810386 PMCID: PMC9944155 DOI: 10.3390/biomimetics8010055] [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: 11/29/2022] [Revised: 01/17/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
Biomaterial research has led to revolutionary healthcare advances. Natural biological macromolecules can impact high-performance, multipurpose materials. This has prompted the quest for affordable healthcare solutions, with a focus on renewable biomaterials with a wide variety of applications and ecologically friendly techniques. Imitating their chemical compositions and hierarchical structures, bioinspired based materials have elevated rapidly over the past few decades. Bio-inspired strategies entail extracting fundamental components and reassembling them into programmable biomaterials. This method may improve its processability and modifiability, allowing it to meet the biological application criteria. Silk is a desirable biosourced raw material due to its high mechanical properties, flexibility, bioactive component sequestration, controlled biodegradability, remarkable biocompatibility, and inexpensiveness. Silk regulates temporo-spatial, biochemical and biophysical reactions. Extracellular biophysical factors regulate cellular destiny dynamically. This review examines the bioinspired structural and functional properties of silk material based scaffolds. We explored silk types, chemical composition, architecture, mechanical properties, topography, and 3D geometry to unlock the body's innate regenerative potential, keeping in mind the novel biophysical properties of silk in film, fiber, and other potential forms, coupled with facile chemical changes, and its ability to match functional requirements for specific tissues.
Collapse
Affiliation(s)
- Ajay Kumar Sahi
- Faculty of Chemistry, Nicolaus Copernicus University in Torun, Jurija Gagarina 11, 87-100 Toruń, Poland
- Correspondence: (A.K.S.); (A.S.)
| | - Shravanya Gundu
- Indian Institute of Technology, School of Biomedical Engineering, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Pooja Kumari
- Indian Institute of Technology, School of Biomedical Engineering, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Tomasz Klepka
- Department of Technology and Polymer Processing, Faculty of Mechanical Engineering, Lublin University of Technology, 36, Nadbystrzycka Str, 20-618 Lublin, Poland
| | - Alina Sionkowska
- Faculty of Chemistry, Nicolaus Copernicus University in Torun, Jurija Gagarina 11, 87-100 Toruń, Poland
- Calisia University, Nowy Świat 4, 62-800 Kalisz, Poland
- Correspondence: (A.K.S.); (A.S.)
| |
Collapse
|
9
|
Huang X, Zeng J, Wang Y. Comparison of the enhanced attachment and proliferation of the human mesenchymal stem cells on the biomimetic nanopatterned surfaces of zein, silk fibroin, and gelatin. J Biomed Mater Res B Appl Biomater 2023; 111:161-172. [PMID: 35906959 DOI: 10.1002/jbm.b.35142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/25/2022] [Accepted: 07/17/2022] [Indexed: 11/10/2022]
Abstract
Natural proteins have been reported to positively affect the attachment and proliferation of cells. For the first time, zein, a plant protein, was utilized to make patterned surface mimicking the extracellular matrix to assist the attachment and proliferation of stem cells. Zein would promote the attachment and proliferation of the stem cells more than 10 times of that of gelatin and silk fibroin, respectively, which are popular protein selections for the formation of the biomaterial scaffolds. The more the surface was covered by zein, the more the stem cell grown. It was revealed that the stem cells would grow and stretch in the direction of the patterns, and the stem cells preferred to grow in the grooves in the size of 8 μm, that was similar to the size of the stem cells, rather than the size larger or smaller than that of the cells, such as 50 and 2 μm. It was concluded that zein is a better choice than silk fibroin and gelatin with highly potential for the formation of patterned surface and structure as the biomaterial scaffolds for stem cell therapy.
Collapse
Affiliation(s)
- Xueying Huang
- Department of Chemistry, Hong Kong Baptist University, Kowloon, Hong Kong
| | - Jie Zeng
- Department of Chemistry, Hong Kong Baptist University, Kowloon, Hong Kong
| | - Yi Wang
- Department of Chemistry, Hong Kong Baptist University, Kowloon, Hong Kong
| |
Collapse
|
10
|
Silk fibroin based interpenetrating network hydrogel for corneal stromal regeneration. Int J Biol Macromol 2022; 223:583-594. [PMID: 36356877 DOI: 10.1016/j.ijbiomac.2022.11.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
There is a need to develop tissue engineering based approaches to address the shortage of donor corneas worldwide for transplantation. To do this a novel approach to fabricate three-dimensional hydrogels using free-radical polymerization was investigated to generate constructs for corneal stromal tissue regeneration. Different ratios of silk fibroin (SF) to polyacrylamide (PA) were used to fabricate semi-interpenetrating hydrogels. Scanning electron micrograph displayed the interconnectivity of pores within the fabricated hydrogels. Pore sizes ranged from 25 to 66 μm. Scaffolds with increasing concentration of SF had enhanced β-sheet structure (verified by Fourier transform infrared spectroscopy). The biological response of human corneal stromal cells to these hydrogels was examined using cellular adhesion, proliferation, cytoskeleton organization, gene expression and immunocytochemical analysis. The fabricated hydrogels possess rapid gelation (∼3 min) at 37 °C, 84 % porosity facilitating keratocyte migration during healing, improved cellular adhesion and no cytotoxicity, indicating their efficiency for in-situ corneal tissue regeneration. Presence of SF in semi-interpenetrating network hydrogel enhanced cellular proliferation, elevated GAG deposition, and increased expression of keratocyte genes, normally associated with healthy corneal stromal tissue. This study acts as an initial step towards fabricating SF based semi-interpenetrating network hydrogels for developing clinically applicable ocular implants.
Collapse
|
11
|
Promising Role of Silk-Based Biomaterials for Ocular-Based Drug Delivery and Tissue Engineering. Polymers (Basel) 2022; 14:polym14245475. [PMID: 36559842 PMCID: PMC9788421 DOI: 10.3390/polym14245475] [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: 03/09/2022] [Revised: 11/14/2022] [Accepted: 12/08/2022] [Indexed: 12/16/2022] Open
Abstract
Silk is a wonderful biopolymer that has a long history of medical applications. Surgical cords and medically authorised human analogues made of silk have a long history of use in management. We describe the use of silk in the treatment of eye diseases in this review by looking at the usage of silk fibroin for eye-related drug delivery applications and medication transfer to the eyes. During this ancient art endeavour, a reduced engineering project that employed silk as a platform for medicine delivery or a cell-filled matrix helped reignite interest. With considerable attention, this study explores the present usage of silk in ocular-based drug delivery. This paper also examines emerging developments with the use of silk as a biopolymer for the treatment of eye ailments. As treatment options for glaucoma, diabetic retinopathy, retinitis pigmentosa, and other retinal diseases and degenerations are developed, the trans-scleral route of drug delivery holds great promise for the selective, sustained-release delivery of these novel therapeutic compounds. We should expect a swarm of silk-inspired materials to enter clinical testing and use on the surface as the secrets of silk are unveiled. This article finishes with a discussion on potential silk power, which adds to better ideas and enhanced ocular medicine delivery.
Collapse
|
12
|
Pourjabbar B, Biazar E, Heidari Keshel S, Baradaran‐Rafii A. Improving the properties of fish skin collagen/silk fibroin dressing by chemical treatment for corneal wound healing. Int Wound J 2022; 20:484-498. [PMID: 35912793 PMCID: PMC9885469 DOI: 10.1111/iwj.13896] [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: 06/01/2022] [Revised: 06/29/2022] [Accepted: 07/04/2022] [Indexed: 02/03/2023] Open
Abstract
Natural biomaterials are crucial in ocular tissue engineering because they allow cells to proliferate, differentiate, and stratify while maintaining the typical epithelial phenotype. In this study, membranes as dressings were formed from silk fibroin and collagen (Co) extracted from fish skin and then modified with carbodiimide chemical cross linker in different concentrations. The samples were evaluated by different analyses such as structural, physical (optical, swelling, denaturation temperature, degradation), mechanical, and biological (viability, cell adhesion, immunocytochemistry) assays. The results showed that all membranes have excellent transparency, especially with higher silk fibroin content. Increasing the cross linker concentration and the ratio of silk fibroin to Co increased the denaturation temperature and mechanical strength and, conversely, reduced the degradation rate and cell adhesion. The samples did not show a significant difference in toxicity with increasing cross linker and silk fibroin ratio. In general, samples with a low silk fibroin ratio combined with cross linker can provide desirable properties as a membrane for corneal wound healing.
Collapse
Affiliation(s)
- Bahareh Pourjabbar
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Esmaeil Biazar
- Tissue Engineering Group, Department of Biomedical Engineering, Tonekabon BranchIslamic Azad UniversityTonekabonIran
| | - Saeed Heidari Keshel
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in MedicineShahid Beheshti University of Medical SciencesTehranIran,Medical Nanotechnology and Tissue Engineering Research CenterShahid Beheshti University of Medical SciencesTehranIran
| | - Alireza Baradaran‐Rafii
- Ophthalmic Research Center, Department of Ophthalmology, Labbafinejad Medical CenterShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
13
|
Kumar A, Yun H, Funderburgh ML, Du Y. Regenerative therapy for the Cornea. Prog Retin Eye Res 2022; 87:101011. [PMID: 34530154 PMCID: PMC8918435 DOI: 10.1016/j.preteyeres.2021.101011] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/13/2022]
Abstract
The cornea is the outmost layer of the eye, unique in its transparency and strength. The cornea not only transmits the light essential for vision, also refracts light, giving focus to images. Each of the three layers of the cornea has properties essential for the function of vision. Although the epithelium can often recover from injury quickly by cell division, loss of limbal stem cells can cause severe corneal surface abnormalities leading to corneal blindness. Disruption of the stromal extracellular matrix and loss of cells determining this structure, the keratocytes, leads to corneal opacity. Corneal endothelium is the inner part of the cornea without self-renewal capacity. It is very important to maintain corneal dehydration and transparency. Permanent damage to the corneal stroma or endothelium can be effectively treated by corneal transplantation; however, there are drawbacks to this procedure, including a shortage of donors, the need for continuing treatment to prevent rejection, and limits to the survival of the graft, averaging 10-20 years. There exists a need for new strategies to promote regeneration of the stromal structure and restore vision. This review highlights critical contributions in regenerative medicine with the aim of corneal reconstruction after injury or disease. These approaches include corneal stromal stem cells, corneal limbal stem cells, embryonic stem cells, and other adult stem cells, as well as induced pluripotent stem cells. Stem cell-derived trophic factors in the forms of secretomes or exosomes for corneal regeneration are also discussed. Corneal sensory nerve regeneration promoting corneal transparency is discussed. This article provides description of the up-to-date options for corneal regeneration and presents exciting possible avenues for future studies toward clinical applications for corneal regeneration.
Collapse
Affiliation(s)
- Ajay Kumar
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA 15213
| | - Hongmin Yun
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA 15213
| | | | - Yiqin Du
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
14
|
Sonnleitner D, Sommer C, Scheibel T, Lang G. Approaches to inhibit biofilm formation applying natural and artificial silk-based materials. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112458. [PMID: 34857315 DOI: 10.1016/j.msec.2021.112458] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022]
Abstract
The discovery of penicillin started a new era of health care since it allowed the effective treatment of formerly deadly infections. As a drawback, its overuse led to a growing number of multi-drug resistant pathogens. Challenging this arising threat, material research focuses on the development of microbe-killing or microbe repellent agents implementing such functions directly into materials. Due to their biocompatibility, non-immunogenicity and mechanical strength, silk-based materials are attractive candidates for applications in the biomedical field. Furthermore, it has been observed that silks display high persistency in their natural environment giving reason to suspect that they might be attractive candidates to prevent microbial infestation. The current review describes the process of biofilm formation on medical devices and the most common strategies to prevent it, divided into effects of surface topography, material modification and integrated additives. In this context, recent state of the art developments in the field of natural and artificial silk-based materials with microbe-repellant or antimicrobial properties are addressed. These silk properties are controversially discussed and conclusions are drawn as to which parameters will be decisive for the successful design of new bio-functional materials based on the blueprint of silk proteins.
Collapse
Affiliation(s)
- David Sonnleitner
- Biopolymer Processing, Faculty of Engineering Science, University of Bayreuth, Germany
| | - Christoph Sommer
- Chair of Biomaterials, Faculty of Engineering Science, University of Bayreuth, Germany
| | - Thomas Scheibel
- Chair of Biomaterials, Faculty of Engineering Science, University of Bayreuth, Germany
| | - Gregor Lang
- Biopolymer Processing, Faculty of Engineering Science, University of Bayreuth, Germany.
| |
Collapse
|
15
|
Formisano N, van der Putten C, Grant R, Sahin G, Truckenmüller RK, Bouten CVC, Kurniawan NA, Giselbrecht S. Mechanical Properties of Bioengineered Corneal Stroma. Adv Healthc Mater 2021; 10:e2100972. [PMID: 34369098 PMCID: PMC11468718 DOI: 10.1002/adhm.202100972] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/15/2021] [Indexed: 12/26/2022]
Abstract
For the majority of patients with severe corneal injury or disease, corneal transplantation is the only suitable treatment option. Unfortunately, the demand for donor corneas greatly exceeds the availability. To overcome shortage issues, a myriad of bioengineered constructs have been developed as mimetics of the corneal stroma over the last few decades. Despite the sheer number of bioengineered stromas developed , these implants fail clinical trials exhibiting poor tissue integration and adverse effects in vivo. Such shortcomings can partially be ascribed to poor biomechanical performance. In this review, existing approaches for bioengineering corneal stromal constructs and their mechanical properties are described. The information collected in this review can be used to critically analyze the biomechanical properties of future stromal constructs, which are often overlooked, but can determine the failure or success of corresponding implants.
Collapse
Affiliation(s)
- Nello Formisano
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229 ERThe Netherlands
| | - Cas van der Putten
- Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5612 APThe Netherlands
| | - Rhiannon Grant
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229 ERThe Netherlands
| | - Gozde Sahin
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229 ERThe Netherlands
| | - Roman K. Truckenmüller
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229 ERThe Netherlands
| | - Carlijn V. C. Bouten
- Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5612 APThe Netherlands
| | - Nicholas A. Kurniawan
- Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5612 APThe Netherlands
| | - Stefan Giselbrecht
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229 ERThe Netherlands
| |
Collapse
|
16
|
Farasatkia A, Kharaziha M. Robust and double-layer micro-patterned bioadhesive based on silk nanofibril/GelMA-alginate for stroma tissue engineering. Int J Biol Macromol 2021; 183:1013-1025. [PMID: 33974922 DOI: 10.1016/j.ijbiomac.2021.05.048] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/25/2021] [Accepted: 05/06/2021] [Indexed: 12/12/2022]
Abstract
We develop a robust micro-patterned double-layer film that can adhere firmly to the tissue and provide a sustained release of ascorbic acid (AA) for corneal regeneration. This double-layer film consists of a AA reservoir sodium alginate (SA) adhesive and an anisotropic layer made of micro-patterned silk nanofibrils (SNF) incorporated gelatin methacrylate (GelMA) (S/G). The S/G layer facilitates the adhesion and orientation of corneal stroma cells, depending on the pattern sizes (50 μm (P1) and 100 (P2) μm). Results reveal that more than 90% and 80% of the cells are located at angles close to the vertical axis (0-20°) in the sample with the smaller and larger pattern size, respectively. The mechanical robustness and 90% light transmission of this hybrid film originate from the micro-patterned S/G layer. However, the micro-pattern size does not show a significant role in the mechanical properties of hybrid films (tensile strength of S/G-SA, S/G-SA(P1), and S/G-SA(P2) is 3.4 ± 0.1 MPa, 3.6 ± 0.6 MPa and 3.3 ± 0.2 MPa, respectively). In addition, the strong adhesion to the tissue of this double-layer film is related to the alginate layer. AA can release in a controlled manner, which can significantly promote corneal stroma cells' attachment, alignment, and proliferation compared to the control (AA-free micro-patterned film). Our results reveal that this innovative multifunctional S/G-SA + AA film can be a proper candidate for use in stroma tissue engineering of the human cornea.
Collapse
Affiliation(s)
- Asal Farasatkia
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Mahshid Kharaziha
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran.
| |
Collapse
|
17
|
Hull SM, Lindsay CD, Brunel LG, Shiwarski DJ, Tashman JW, Roth JG, Myung D, Feinberg AW, Heilshorn SC. 3D Bioprinting using UNIversal Orthogonal Network (UNION) Bioinks. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2007983. [PMID: 33613150 PMCID: PMC7888563 DOI: 10.1002/adfm.202007983] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Indexed: 05/02/2023]
Abstract
Three-dimensional (3D) bioprinting is a promising technology to produce tissue-like structures, but a lack of diversity in bioinks is a major limitation. Ideally each cell type would be printed in its own customizable bioink. To fulfill this need for a universally applicable bioink strategy, we developed a versatile, bioorthogonal bioink crosslinking mechanism that is cell compatible and works with a range of polymers. We term this family of materials UNIversal, Orthogonal Network (UNION) bioinks. As demonstration of UNION bioink versatility, gelatin, hyaluronic acid (HA), recombinant elastin-like protein (ELP), and polyethylene glycol (PEG) were each used as backbone polymers to create inks with storage moduli spanning 200 to 10,000 Pa. Because UNION bioinks are crosslinked by a common chemistry, multiple materials can be printed together to form a unified, cohesive structure. This approach is compatible with any support bath that enables diffusion of UNION crosslinkers. Both matrix-adherent human corneal mesenchymal stromal cells and non-matrix-adherent human induced pluripotent stem cell-derived neural progenitor spheroids were printed with UNION bioinks. The cells retained high viability and expressed characteristic phenotypic markers after printing. Thus, UNION bioinks are a versatile strategy to expand the toolkit of customizable materials available for 3D bioprinting.
Collapse
Affiliation(s)
- Sarah M Hull
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Christopher D Lindsay
- Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA
| | - Lucia G Brunel
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Daniel J Shiwarski
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Joshua W Tashman
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Julien G Roth
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - David Myung
- Department of Ophthalmology, Stanford University, Stanford, CA 94305, USA
| | - Adam W Feinberg
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Sarah C Heilshorn
- Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
18
|
Guérin LP, Le-Bel G, Desjardins P, Couture C, Gillard E, Boisselier É, Bazin R, Germain L, Guérin SL. The Human Tissue-Engineered Cornea (hTEC): Recent Progress. Int J Mol Sci 2021; 22:ijms22031291. [PMID: 33525484 PMCID: PMC7865732 DOI: 10.3390/ijms22031291] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/11/2022] Open
Abstract
Each day, about 2000 U.S. workers have a job-related eye injury requiring medical treatment. Corneal diseases are the fifth cause of blindness worldwide. Most of these diseases can be cured using one form or another of corneal transplantation, which is the most successful transplantation in humans. In 2012, it was estimated that 12.7 million people were waiting for a corneal transplantation worldwide. Unfortunately, only 1 in 70 patients received a corneal graft that same year. In order to provide alternatives to the shortage of graftable corneas, considerable progress has been achieved in the development of living corneal substitutes produced by tissue engineering and designed to mimic their in vivo counterpart in terms of cell phenotype and tissue architecture. Most of these substitutes use synthetic biomaterials combined with immortalized cells, which makes them dissimilar from the native cornea. However, studies have emerged that describe the production of tridimensional (3D) tissue-engineered corneas using untransformed human corneal epithelial cells grown on a totally natural stroma synthesized by living corneal fibroblasts, that also show appropriate histology and expression of both extracellular matrix (ECM) components and integrins. This review highlights contributions from laboratories working on the production of human tissue-engineered corneas (hTECs) as future substitutes for grafting purposes. It overviews alternative models to the grafting of cadaveric corneas where cell organization is provided by the substrate, and then focuses on their 3D counterparts that are closer to the native human corneal architecture because of their tissue development and cell arrangement properties. These completely biological hTECs are therefore very promising as models that may help understand many aspects of the molecular and cellular mechanistic response of the cornea toward different types of diseases or wounds, as well as assist in the development of novel drugs that might be promising for therapeutic purposes.
Collapse
Affiliation(s)
- Louis-Philippe Guérin
- CUO-Recherche, Médecine Régénératrice—Centre de Recherche du CHU de Québec, Université Laval, Québec, QC G1S 4L8, Canada; (L.-P.G.); (G.L.-B.); (P.D.); (C.C.); (E.G.); (É.B.); (R.B.); (L.G.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Québec, QC G1J 1Z4, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Gaëtan Le-Bel
- CUO-Recherche, Médecine Régénératrice—Centre de Recherche du CHU de Québec, Université Laval, Québec, QC G1S 4L8, Canada; (L.-P.G.); (G.L.-B.); (P.D.); (C.C.); (E.G.); (É.B.); (R.B.); (L.G.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Québec, QC G1J 1Z4, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Département de Chirurgie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Pascale Desjardins
- CUO-Recherche, Médecine Régénératrice—Centre de Recherche du CHU de Québec, Université Laval, Québec, QC G1S 4L8, Canada; (L.-P.G.); (G.L.-B.); (P.D.); (C.C.); (E.G.); (É.B.); (R.B.); (L.G.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Québec, QC G1J 1Z4, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Département de Chirurgie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Camille Couture
- CUO-Recherche, Médecine Régénératrice—Centre de Recherche du CHU de Québec, Université Laval, Québec, QC G1S 4L8, Canada; (L.-P.G.); (G.L.-B.); (P.D.); (C.C.); (E.G.); (É.B.); (R.B.); (L.G.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Québec, QC G1J 1Z4, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Département de Chirurgie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Elodie Gillard
- CUO-Recherche, Médecine Régénératrice—Centre de Recherche du CHU de Québec, Université Laval, Québec, QC G1S 4L8, Canada; (L.-P.G.); (G.L.-B.); (P.D.); (C.C.); (E.G.); (É.B.); (R.B.); (L.G.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Québec, QC G1J 1Z4, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Élodie Boisselier
- CUO-Recherche, Médecine Régénératrice—Centre de Recherche du CHU de Québec, Université Laval, Québec, QC G1S 4L8, Canada; (L.-P.G.); (G.L.-B.); (P.D.); (C.C.); (E.G.); (É.B.); (R.B.); (L.G.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Québec, QC G1J 1Z4, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Richard Bazin
- CUO-Recherche, Médecine Régénératrice—Centre de Recherche du CHU de Québec, Université Laval, Québec, QC G1S 4L8, Canada; (L.-P.G.); (G.L.-B.); (P.D.); (C.C.); (E.G.); (É.B.); (R.B.); (L.G.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Québec, QC G1J 1Z4, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Lucie Germain
- CUO-Recherche, Médecine Régénératrice—Centre de Recherche du CHU de Québec, Université Laval, Québec, QC G1S 4L8, Canada; (L.-P.G.); (G.L.-B.); (P.D.); (C.C.); (E.G.); (É.B.); (R.B.); (L.G.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Québec, QC G1J 1Z4, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Département de Chirurgie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Sylvain L. Guérin
- CUO-Recherche, Médecine Régénératrice—Centre de Recherche du CHU de Québec, Université Laval, Québec, QC G1S 4L8, Canada; (L.-P.G.); (G.L.-B.); (P.D.); (C.C.); (E.G.); (É.B.); (R.B.); (L.G.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Québec, QC G1J 1Z4, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Correspondence: ; Tel.: +1-418-682-7565
| |
Collapse
|
19
|
Karayilan M, Clamen L, Becker ML. Polymeric Materials for Eye Surface and Intraocular Applications. Biomacromolecules 2021; 22:223-261. [PMID: 33405900 DOI: 10.1021/acs.biomac.0c01525] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Ocular applications of polymeric materials have been widely investigated for medical diagnostics, treatment, and vision improvement. The human eye is a vital organ that connects us to the outside world so when the eye is injured, infected, or impaired, it needs immediate medical treatment to maintain clear vision and quality of life. Moreover, several essential parts of the eye lose their functions upon aging, causing diminished vision. Modern polymer science and polymeric materials offer various alternatives, such as corneal and scleral implants, artificial ocular lenses, and vitreous substitutes, to replace the damaged parts of the eye. In addition to the use of polymers for medical treatment, polymeric contact lenses can provide not only vision correction, but they can also be used as wearable electronics. In this Review, we highlight the evolution of polymeric materials for specific ocular applications such as intraocular lenses and current state-of-the-art polymeric systems with unique properties for contact lens, corneal, scleral, and vitreous body applications. We organize this Review paper by following the path of light as it travels through the eye. Starting from the outside of the eye (contact lenses), we move onto the eye's surface (cornea and sclera) and conclude with intraocular applications (intraocular lens and vitreous body) of mostly synthetic polymers and several biopolymers. Initially, we briefly describe the anatomy and physiology of the eye as a reminder of the eye parts and their functions. The rest of the Review provides an overview of recent advancements in next-generation contact lenses and contact lens sensors, corneal and scleral implants, solid and injectable intraocular lenses, and artificial vitreous body. Current limitations for future improvements are also briefly discussed.
Collapse
Affiliation(s)
- Metin Karayilan
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Liane Clamen
- Adaptilens, LLC, Boston, Massachusetts 02467, United States
| | - Matthew L Becker
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States.,Mechanical Engineering and Materials Science, Orthopaedic Surgery, and Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| |
Collapse
|
20
|
Pollard RE, McKay TB, Ford A, Cairns DM, Georgakoudi I, Kaplan DL. Induction of Irritation and Inflammation in a 3D Innervated Tissue Model of the Human Cornea. ACS Biomater Sci Eng 2020; 6:6886-6895. [PMID: 33320598 DOI: 10.1021/acsbiomaterials.0c01136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Detection of slight changes in the chemical, thermal, and physical environments of the ocular surface is necessary to protect eyesight. The cornea, as the most densely innervated peripheral tissue in the body, can be damaged as a result of caustic chemical exposure. Such damage can be painful and debilitating, thus underscoring the need to understand mechanisms of ocular irritation. Both ethical and translational limitations regarding the use of animal subjects in part drive the need to develop relevant in vitro cell and tissue models that emulate the physiology of the human cornea. In this study, we utilized our 3D in vitro cornea-like tissue model to study the effects of irritation mediated by transient receptor potential (TRP) channels vanilloid 1 and ankyrin 1 (TRPV1; TRPA1) in response to allyl isothiocyanate (AITC) stimulation. Changes in gene expression were analyzed to characterize wound healing responses of the epithelial, stromal, and neuronal cell populations in the corneal tissue models. Key findings of the study include indications of wound healing, such as stromal myofibroblast differentiation and epithelial barrier re-establishment, amplification of pro-inflammatory cytokines, and downstream ECM protein remodeling due to irritation with the addition of sensory innervation. This study further establishes this in vitro tissue model as a useful tool for studying corneal irritation in vitro in a holistic manner with promise as a novel and sensitive tool for studying chemical exposures and subsequent responses.
Collapse
Affiliation(s)
- Rachel E Pollard
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, Massachusetts 02155, United States
| | - Tina B McKay
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, Massachusetts 02155, United States
| | - Andrew Ford
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, Massachusetts 02155, United States
| | - Dana M Cairns
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, Massachusetts 02155, United States
| | - Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, Massachusetts 02155, United States
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, Massachusetts 02155, United States
| |
Collapse
|
21
|
Bionic Silk Fibroin Film Promotes Tenogenic Differentiation of Tendon Stem/Progenitor Cells by Activating Focal Adhesion Kinase. Stem Cells Int 2020; 2020:8857380. [PMID: 33204279 PMCID: PMC7657703 DOI: 10.1155/2020/8857380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 10/10/2020] [Accepted: 10/17/2020] [Indexed: 01/08/2023] Open
Abstract
Background Tendon injuries are common musculoskeletal disorders in clinic. Due to the limited regeneration ability of tendons, tissue engineering technology is often used as an effective approach to treat tendon injuries. Silk fibroin (SF) films have excellent biological activities and physical properties, which is suitable for tendon regeneration. The present study is aimed at preparing a SF film with a bionic microstructure and investigating its biological effects. Methods A SF film with a smooth surface or bionic microstructure was prepared. After seeding tendon stem/progenitor cells (TSPCs) on the surface, the cell morphology, the expression level of tenogenic genes and proteins, and the focal adhesion kinase (FAK) activation were measured to evaluate the biological effect of SF films. Results The TSPCs on SF films with a bionic microstructure exhibited a slender cell morphology, promoted the expression of tenogenic genes and proteins, such as SCX, TNC, TNMD, and COLIA1, and activated FAK. FAK inhibitors blocked the enhanced expression of tenogenic genes and proteins. Conclusion SF films with a bionic microstructure may serve as a scaffold, provide biophysical cues to alter the cellular adherence arrangement and cell morphology, and enhance the tenogenic gene and protein expression in TSPCs. FAK activation plays a key role during this biological response process.
Collapse
|
22
|
Madden PW, Klyubin I, Ahearne MJ. Silk fibroin safety in the eye: a review that highlights a concern. BMJ Open Ophthalmol 2020; 5:e000510. [PMID: 33024827 PMCID: PMC7513638 DOI: 10.1136/bmjophth-2020-000510] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/15/2020] [Accepted: 08/07/2020] [Indexed: 12/25/2022] Open
Abstract
The biomedical use of silk as a suture dates back to antiquity. Fibroin is the structural element that determines the strength of silk and here we consider the safety of fibroin in its role in ophthalmology. The high mechanical strength of silk meant sufficiently thin threads could be made for eye microsurgery, but such usage was all but superseded by synthetic polymer sutures, primarily because silk in its entirety was more inflammatory. Significant immunological response can normally be avoided by careful manufacturing to provide high purity fibroin, and it has been utilised in this form for tissue engineering an array of fibre and film substrata deployed in research with cells of the eye. Films of fibroin can also be made transparent, which is a required property in the visual pathway. Transparent layers of corneal epithelial, stromal and endothelial cells have all been demonstrated with maintenance of phenotype, as have constructs supporting retinal cells. Fibroin has a lack of demonstrable infectious agent transfer, an ability to be sterilised and prepared with minimal contamination, long-term predictable degradation and low direct cytotoxicity. However, there remains a known ability to be involved in amyloid formation and potential amyloidosis which, without further examination, is enough to currently question whether fibroin should be employed in the eye given its innervation into the brain.
Collapse
Affiliation(s)
- Peter W Madden
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, the University of Dublin, Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, the University of Dublin, Dublin, Ireland
| | - Igor Klyubin
- Department of Pharmacology Therapeutics, School of Medicine, Trinity College Dublin, the University of Dublin, Dublin, Ireland
- Institute of Neuroscience, Trinity College Dublin, the University of Dublin, Dublin, Ireland
| | - Mark J Ahearne
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, the University of Dublin, Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, the University of Dublin, Dublin, Ireland
| |
Collapse
|
23
|
Lau K, Akhavan B, Lord MS, Bilek MM, Rnjak-Kovacina J. Dry Surface Treatments of Silk Biomaterials and Their Utility in Biomedical Applications. ACS Biomater Sci Eng 2020; 6:5431-5452. [PMID: 33320554 DOI: 10.1021/acsbiomaterials.0c00888] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Silk-based materials are widely used in biomaterial and tissue engineering applications due to their cytocompatibility and tunable mechanical and biodegradation properties. Aqueous-based processing techniques have enabled the fabrication of silk into a broad range of material formats, making it a highly versatile material platform across multiple industries. Utilizing the full potential of silk in biomedical applications frequently requires modification of silk's surface properties. Dry surface modification techniques, including irradiation and plasma treatment, offer an alternative to the conventional wet chemistry strategies to modify the physical and chemical properties of silk materials without compromising their bulk properties. While dry surface modification techniques are more prevalent in textiles and sterilization applications, the range of modifications available and resultant changes to silk materials all point to the utility of dry surface modification for the development of new, functional silk biomaterials. Dry surface treatment affects the surface chemistry, secondary structure, molecular weight, topography, surface energy, and mechanical properties of silk materials. This Review describes and critically evaluates the effect of physical dry surface modification techniques, including irradiation and plasma processes, on silk materials and discusses their utility in biomedical applications, including recent examples of modulation of cell/protein interactions on silk biomaterials, in vivo performance of implanted biomaterials, and applications in material biofunctionalization and lithographic surface patterning approaches.
Collapse
Affiliation(s)
- Kieran Lau
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Behnam Akhavan
- School of Physics, University of Sydney, Sydney, NSW 2006, Australia.,School of Biomedical Engineering, University of Sydney, Sydney, NSW 2006, Australia.,University of Sydney Nano Institute, University of Sydney, Sydney NSW 2006, Australia
| | - Megan S Lord
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Marcela M Bilek
- School of Physics, University of Sydney, Sydney, NSW 2006, Australia.,School of Biomedical Engineering, University of Sydney, Sydney, NSW 2006, Australia.,University of Sydney Nano Institute, University of Sydney, Sydney NSW 2006, Australia.,Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
24
|
McKay TB, Hutcheon AEK, Guo X, Zieske JD, Karamichos D. Modeling the cornea in 3-dimensions: Current and future perspectives. Exp Eye Res 2020; 197:108127. [PMID: 32619578 PMCID: PMC8116933 DOI: 10.1016/j.exer.2020.108127] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 06/17/2020] [Accepted: 06/23/2020] [Indexed: 02/08/2023]
Abstract
The cornea is an avascular, transparent ocular tissue that serves as a refractive and protective structure for the eye. Over 90% of the cornea is composed of a collagenous-rich extracellular matrix within the stroma with the other 10% composed by the corneal epithelium and endothelium layers and their corresponding supporting collagen layers (e.g., Bowman's and Descemet's membranes) at the anterior and posterior cornea, respectively. Due to its prominent role in corneal structure, tissue engineering approaches to model the human cornea in vitro have focused heavily on the cellular and functional properties of the corneal stroma. In this review, we discuss model development in the context of culture dimensionality (e.g., 2-dimensional versus 3-dimensional) and expand on the optical, biomechanical, and cellular functions promoted by the culture microenvironment. We describe current methods to model the human cornea with focus on organotypic approaches, compressed collagen, bioprinting, and self-assembled stromal models. We also expand on co-culture applications with the inclusion of relevant corneal cell types, such as epithelial, stromal keratocyte or fibroblast, endothelial, and neuronal cells. Further advancements in corneal tissue model development will markedly improve our current understanding of corneal wound healing and regeneration.
Collapse
Affiliation(s)
- Tina B McKay
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Audrey E K Hutcheon
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Xiaoqing Guo
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - James D Zieske
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Dimitrios Karamichos
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA.
| |
Collapse
|
25
|
McKay TB, Ford A, Wang S, Cairns DM, Parker RN, Deardorff PM, Ghezzi CE, Kaplan DL. Assembly and Application of a Three-Dimensional Human Corneal Tissue Model. ACTA ACUST UNITED AC 2020; 81:e84. [PMID: 31529796 DOI: 10.1002/cptx.84] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The cornea provides a functional barrier separating the outside environment from the intraocular environment, thereby protecting posterior segments of the eye from infection and damage. Pathological changes that compromise the structure or integrity of the cornea may occur as a result of injury or disease and can lead to debilitating effects on visual acuity. Over 10 million people worldwide are visually impaired or blind due to corneal opacity. Thus, physiologically relevant in vitro approaches to predict corneal toxicity of chemicals or effective treatments for disease prior to ocular exposure, as well as to study the corneal effects of systemic, chronic conditions, such as diabetes, are needed to reduce use of animal testing and accelerate therapeutic development. We have previously bioengineered an innervated corneal tissue model using silk protein scaffolds to recapitulate the structural and mechanical elements of the anterior cornea and to model the functional aspects of corneal sensation with the inclusion of epithelial, stromal, and neural components. The purpose of this unit is to provide a step-by-step guide for preparation, assembly, and application of this three-dimensional corneal tissue system to enable the study of corneal tissue biology. © 2019 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Tina B McKay
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Andrew Ford
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Siran Wang
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Dana M Cairns
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Rachael N Parker
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Phillip M Deardorff
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Chiara E Ghezzi
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| |
Collapse
|
26
|
Wang K, Man K, Liu J, Liu Y, Chen Q, Zhou Y, Yang Y. Microphysiological Systems: Design, Fabrication, and Applications. ACS Biomater Sci Eng 2020; 6:3231-3257. [PMID: 33204830 PMCID: PMC7668566 DOI: 10.1021/acsbiomaterials.9b01667] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Microphysiological systems, including organoids, 3-D printed tissue constructs and organ-on-a-chips (organ chips), are physiologically relevant in vitro models and have experienced explosive growth in the past decades. Different from conventional, tissue culture plastic-based in vitro models or animal models, microphysiological systems recapitulate key microenvironmental characteristics of human organs and mimic their primary functions. The advent of microphysiological systems is attributed to evolving biomaterials, micro-/nanotechnologies and stem cell biology, which enable the precise control over the matrix properties and the interactions between cells, tissues and organs in physiological conditions. As such, microphysiological systems have been developed to model a broad spectrum of organs from microvasculature, eye, to lung and many others to understand human organ development and disease pathology and facilitate drug discovery. Multiorgans-on-a-chip systems have also been developed by integrating multiple associated organ chips in a single platform, which allows to study and employ the organ function in a systematic approach. Here we first discuss the design principles of microphysiological systems with a focus on the anatomy and physiology of organs, and then review the commonly used fabrication techniques and biomaterials for microphysiological systems. Subsequently, we discuss the recent development of microphysiological systems, and provide our perspectives on advancing microphysiological systems for preclinical investigation and drug discovery of human disease.
Collapse
Affiliation(s)
- Kai Wang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Kun Man
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Jiafeng Liu
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Yang Liu
- North Texas Eye Research Institute, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas 76107, United States
| | - Qi Chen
- The Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Yong Zhou
- Department of Emergency, Xinqiao Hospital, Chongqing 400037, China
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| |
Collapse
|
27
|
Ramachandran C, Gupta P, Hazra S, Mandal BB. In Vitro Culture of Human Corneal Endothelium on Non-Mulberry Silk Fibroin Films for Tissue Regeneration. Transl Vis Sci Technol 2020; 9:12. [PMID: 32818099 PMCID: PMC7396167 DOI: 10.1167/tvst.9.4.12] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 12/27/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose The purpose of this study was to determine if non-mulberry varieties of silk are suitable for the culture of corneal endothelium (CE). Methods Aqueous silk fibroin derived from Philosamia ricini (PR), Antheraea assamensis (AA), and Bombyx mori (BM) were cast as approximately 15 µm films with and without pores on which human CE cells were cultured. Tensile strength, elasticity, transmittance in visible range, and degradation properties of the films were characterised. Adhesion of CE to the silk films was quantified using MTT assay in addition to quantifying the number and area of focal adhesions using paxillin. Expression of CE markers was determined at the gene and protein levels using PCR and immunostaining, respectively. Barrier integrity of the cultured cells was measured as permeability to FITC dextran (10 kDa) in the presence or absence of thrombin. Results The films exhibited robust tensile strength, >95% transmittance and a refractive index comparable to the native cornea. BM degraded significantly faster when compared to PR and AA. A comparison between the three varieties of silk showed that significantly more cells were adhered to PR and AA than to BM. This was also reflected in the expression of stable focal adhesions on PR and AA, thus enabling the formation of intact monolayers of cells on these varieties unlike on BM. Treatment with thrombin significantly increased cellular permeability to dextran. Conclusions Our data shows that PR and AA varieties sufficiently support the growth and function of CE cells. This could be attributed to the presence of natural cell binding motifs (RGD) in these varieties. Translational Relevance Development of a suitable carrier for engineering the CE to address a major clinical requirement of healthy donor tissues for transplantation.
Collapse
Affiliation(s)
- Charanya Ramachandran
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, Telangana, India
| | - Prerak Gupta
- Biomaterial and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Swatilekha Hazra
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, Telangana, India.,Manipal University, Manipal, India
| | - Biman B Mandal
- Biomaterial and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India.,Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| |
Collapse
|
28
|
Xiong S, Gao H, Qin L, Jia YG, Ren L. Engineering topography: Effects on corneal cell behavior and integration into corneal tissue engineering. Bioact Mater 2019; 4:293-302. [PMID: 31709312 PMCID: PMC6829100 DOI: 10.1016/j.bioactmat.2019.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 08/23/2019] [Accepted: 10/07/2019] [Indexed: 12/13/2022] Open
Abstract
Cell-material interactions are important to tissue engineering. Inspired by the natural topographic structures on the extracellular matrix, a growing number of studies have integrated engineering topography into investigations of cell behavior on biomaterials. Engineering topography has a significant influence on cell behaviors. These cell-topography interactions play an important role in regenerative medicine and tissue engineering. Similarly, cell-topography interactions are important to corneal reconstruction and regeneration. In this review, we primarily summarized the effects of topographic cues on the behaviors of corneal cells, including cell morphology, adhesion, migration, and proliferation. Furthermore, the integration of engineering surface topography into corneal tissue engineering was also discussed.
Collapse
Affiliation(s)
- Sijia Xiong
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, Guangzhou, 510006, China
| | - HuiChang Gao
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Lanfeng Qin
- National Engineering Research Centre for Tissue Restoration and Reconstruction, Guangzhou, 510006, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Yong-Guang Jia
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, Guangzhou, 510006, China
| | - Li Ren
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, Guangzhou, 510006, China
- Sino-Singapore International Joint Research Institute, Guangzhou, 510555, China
| |
Collapse
|
29
|
Potential for combined delivery of riboflavin and all-trans retinoic acid, from silk fibroin for corneal bioengineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 105:110093. [DOI: 10.1016/j.msec.2019.110093] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 08/08/2019] [Accepted: 08/15/2019] [Indexed: 12/20/2022]
|
30
|
Moghanizadeh-Ashkezari M, Shokrollahi P, Zandi M, Shokrolahi F, Daliri MJ, Kanavi MR, Balagholi S. Vitamin C Loaded Poly(urethane-urea)/ZnAl-LDH Aligned Scaffolds Increase Proliferation of Corneal Keratocytes and Up-Regulate Vimentin Secretion. ACS APPLIED MATERIALS & INTERFACES 2019; 11:35525-35539. [PMID: 31490646 DOI: 10.1021/acsami.9b07556] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
A novel poly(urethane-urea) (PUU) based on poly(glycolide-co-ε-caprolactone) macro-diol with tunable mechanical properties and biodegradation behavior is reported for corneal stromal tissue regeneration. Zn-Al layered double hydroxide (LDH) nanoparticles were synthesized and loaded with vitamin C (VC, VC-LDH) and dispersed in the PUU to control VC release in the cell culturing medium. To mimic the corneal stromal EC, scaffolds of the PUU and its nanocomposites with VC-LDH (PUU-LDH and PUU-VC-LDH) were fabricated via electrospinning. Average diameters of the aligned nanofibers were recorded as 325 ± 168, 343 ± 171, and 414 ± 275 nm for the PUU, PUU-LDH, and PUU-VC-LDH scaffolds, respectively. Results of hydrophilicity and mechanical properties measurements showed increased hydrophobicity and reduced tensile strength and elongation at break upon addition of nanoparticles to the PUU scaffold. VC release studies represented that intercalation of the drug in Zn-Al-LDH controlled the burst release and extended the release period from a few hours to 5 days. Viability and proliferation of stromal keratocyte cells on the scaffolds were investigated via AlamarBlue assay. After 24 h, the cells showed similar viability on the scaffolds and the control. After 1 week, the cells showed some degree of proliferation on the scaffolds, with the highest value recorded for PUU-VC-LDH. SEM images of the scaffolds after 24 h and 1 week confirmed good penetration and attachment of keratocytes on all the scaffolds and the cells oriented with the direction of nanofibers. After 1 week, the PUU-VC-LDH scaffold was fully covered by the cells. Immunocytochemistry assay (ICC) was performed to investigate secretion of vimentin protein, ALDH3A1, and α-SMA by the cells. After 24h and 1 week, remarkably higher levels of vimentin and ALDH3A1 and lower level of α-SMA were secreted by keratocytes on PUU-VC-LDH compared to those on the PUU and PUU-LDH scaffolds and the control. Our results suggest that the aligned PUU-VC-LDH is a promising candidate for corneal stromal tissue engineering due to the presence of zinc and vitamin C.
Collapse
Affiliation(s)
- Mojgan Moghanizadeh-Ashkezari
- Department of Biomaterials, Faculty of Science , Iran Polymer and Petrochemical Institute , 14977-13115 , Tehran , Iran
| | - Parvin Shokrollahi
- Department of Biomaterials, Faculty of Science , Iran Polymer and Petrochemical Institute , 14977-13115 , Tehran , Iran
| | - Mojgan Zandi
- Department of Biomaterials, Faculty of Science , Iran Polymer and Petrochemical Institute , 14977-13115 , Tehran , Iran
| | - Fatemeh Shokrolahi
- Department of Biomaterials, Faculty of Science , Iran Polymer and Petrochemical Institute , 14977-13115 , Tehran , Iran
| | - Morteza J Daliri
- Department of Animal and Marine Biotechnology , National Institute of Genetic Engineering and Biotechnology , 14977-16316 Tehran , Iran
| | - Mozhgan R Kanavi
- Ocular Tissue Engineering Research Center , Shahid Beheshti University of Medical Sciences , 16666-63111 , Tehran , Iran
| | - Sahar Balagholi
- Blood Transfusion Research Center , High Institute for Research and Education in Transfusion Medicine , 14665-1157 , Tehran , Iran
| |
Collapse
|
31
|
McKay TB, Parker RN, Hawker MJ, McGill M, Kaplan DL. Silk-Based Therapeutics Targeting Pseudomonas aeruginosa. J Funct Biomater 2019; 10:jfb10030041. [PMID: 31540233 PMCID: PMC6787730 DOI: 10.3390/jfb10030041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 11/16/2022] Open
Abstract
Pseudomonas aeruginosa (P. aeruginosa) infections may lead to severe damage of the cornea, mucosa, and skin. The highly aggressive nature of P. aeruginosa and the rise in multi-drug resistance, particularly in nosocomial settings, lead to an increased risk for permanent tissue damage and potentially death. Thus, a growing need exists to develop alternative treatments to reduce both the occurrence of bacterial infection and biofilm development, as well as pathological progression post-infection. Silk derived from Bombyx mori silkworms serves as a unique biomaterial that is biocompatible with low immunogenicity and high versatility, and thereby ideal for stabilizing therapeutics. In this study, we assessed the cytotoxicity of P. aeruginosa on human corneal stromal stem cells and two mucosal cell lines (Caco-2 and HT29-MTX). To determine whether antibiotic-immobilized scaffolds can serve as alternative therapeutics to free, diffuse forms, we developed novel gentamicin-conjugated silk films as functional scaffolds and compared antimicrobial effects and free gentamicin. The advantages of generating a surface coating with a covalently-bound antibiotic may reduce potential side-effects associated with free gentamicin, as well as limit the diffusion of the drug. Our results suggest that gentamicin conjugated to native silk and carboxyl-enriched silk inhibits P. aeruginosa growth. Development of stabilized antibiotic treatments with surface toxicity selective against bacteria may serve as an alternative approach to treat active infections, as well as potential prophylactic use as coatings in high-risk cases, such as post-surgical complications or prolonged hospitalization.
Collapse
Affiliation(s)
- Tina B McKay
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA.
| | - Rachael N Parker
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA.
| | - Morgan J Hawker
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA.
| | - Meghan McGill
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA.
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA.
| |
Collapse
|
32
|
Che X, Wu H, Jia C, Sun H, Ou S, Wang J, Jeyalatha MV, He X, Yu J, Zuo C, Liu Z, Li W. A Novel Tissue-Engineered Corneal Stromal Equivalent Based on Amniotic Membrane and Keratocytes. Invest Ophthalmol Vis Sci 2019; 60:517-527. [PMID: 30707753 DOI: 10.1167/iovs.18-24869] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To investigate a novel strategy in constructing tissue-engineered corneal stromal equivalent based on amniotic membrane and keratocytes. Methods The ultrathin amniotic membrane (UAM) was laminated, with corneal stromal cells (CSCs) distributed between the space of the layered UAMs. Calcein AM staining was used to evaluate cellular viability, morphology, and arrangement. Immunostaining, qRT-PCR, and Western blot were performed to detect gene and protein expression in keratocytes. Optical coherence tomography visualized the cross sections and thickness of the UAM construction. The microstructure of the CSC-secreted extracellular matrix (ECM) was investigated by scanning electron microscopy and transmission electron microscopy (TEM). To evaluate the feasibility of the multilayer UAM-CSC lamination for surgery, the corneal substitute was used to perform lamellar keratoplasty. Slit lamp microscopy and corneal fluorescein staining were performed in postsurgery observation. Results The CSCs maintained their keratocyte phenotype and secreted well-organized ECM on the aligned UAM surface. The four-layer UAM-CSC lamination attained half thickness of the human cornea (250 ± 18 μm) after 8 weeks' culture, which also showed promising optimal transparency. In TEM images, the CSC-generated ECM displayed stratified, multilayered lamellae with orthogonal fibril arrangement, which was similar to the human cornea microstructure. Furthermore, the stromal equivalent was successfully preformed in lamellar keratoplasty. Four weeks post surgery, the substitute was well integrated into the recipient cornea and completely epithelialized without myofibroblast differentiation. Conclusions Our study established a novel 3D biomimetic corneal model to replicate the corneal stromal organization with multilayer UAM, which was capable of promoting the development of corneal stroma-like tissues in vitro, establishing a new avenue for basic research and therapeutic potential.
Collapse
Affiliation(s)
- Xin Che
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Han Wu
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Changkai Jia
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Huimin Sun
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Shangkun Ou
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Junqi Wang
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - M Vimalin Jeyalatha
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xin He
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jingwen Yu
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Chengyou Zuo
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zuguo Liu
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.,Xiamen University Affiliated Xiamen Eye Center, Xiamen, Fujian, China
| | - Wei Li
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.,Xiamen University Affiliated Xiamen Eye Center, Xiamen, Fujian, China
| |
Collapse
|
33
|
Kim H, Park MN, Kim J, Jang J, Kim HK, Cho DW. Characterization of cornea-specific bioink: high transparency, improved in vivo safety. J Tissue Eng 2019; 10:2041731418823382. [PMID: 30719272 PMCID: PMC6348552 DOI: 10.1177/2041731418823382] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 12/16/2018] [Indexed: 12/22/2022] Open
Abstract
Corneal transplantation is a typical surgical procedure for severe corneal diseases. However, the waiting time for a donor cornea has gradually increased due to a decrease in supply caused by an aging population and increased cases of laser-based surgeries. Artificial corneas were developed to meet the increase in demand; however, these approaches have suffered from material deterioration resulted by the limited tissue integration. Here, we introduce a cornea-derived decellularized extracellular matrix (Co-dECM) as a bioink for corneal regeneration. The developed Co-dECM bioink had similar quantitative measurement results for collagen and GAGs compared with that of the native cornea and also had the proper transparency for vision. The differentiation potential of human turbinate-derived mesenchymal stem cells (hTMSCs) to a keratocyte lineage was only observed in the Co-dECM group. Moreover, the developed bioink did not have any cytotoxic effect on encapsulated cells for three-dimensional (3D) culture and has great biocompatibility evident by the xeno-implantation of the Co-dECM gel into mice and rabbits for two and one month, respectively. An in vivo safety similar to clinical-grade collagen was seen with the Co-dECM, which helped to maintain the keratocyte-specific characteristics in vivo, compared with collagen. Taken together, the Co-dECM bioink has the potential to be used in various types of corneal diseases based on its corneal-specific ability and design flexibility through 3D cell printing technology.
Collapse
Affiliation(s)
- Hyeonji Kim
- Department of Mechanical Engineering and Center for Rapid Prototyping-based 3D Tissue/Organ Printing, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Moon-Nyeo Park
- College of Korean Medicines, Kyung Hee University, Seoul, Republic of Korea
| | - Jisoo Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Jinah Jang
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Republic of Korea
- Department of Creative IT Engineering, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Hong-Kyun Kim
- Department of Ophthalmology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering and Center for Rapid Prototyping-based 3D Tissue/Organ Printing, Pohang University of Science and Technology, Pohang, Republic of Korea
| |
Collapse
|
34
|
Holland C, Numata K, Rnjak‐Kovacina J, Seib FP. The Biomedical Use of Silk: Past, Present, Future. Adv Healthc Mater 2019; 8:e1800465. [PMID: 30238637 DOI: 10.1002/adhm.201800465] [Citation(s) in RCA: 396] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 08/04/2018] [Indexed: 11/07/2022]
Abstract
Humans have long appreciated silk for its lustrous appeal and remarkable physical properties, yet as the mysteries of silk are unraveled, it becomes clear that this outstanding biopolymer is more than a high-tech fiber. This progress report provides a critical but detailed insight into the biomedical use of silk. This journey begins with a historical perspective of silk and its uses, including the long-standing desire to reverse engineer silk. Selected silk structure-function relationships are then examined to appreciate past and current silk challenges. From this, biocompatibility and biodegradation are reviewed with a specific focus of silk performance in humans. The current clinical uses of silk (e.g., sutures, surgical meshes, and fabrics) are discussed, as well as clinical trials (e.g., wound healing, tissue engineering) and emerging biomedical applications of silk across selected formats, such as silk solution, films, scaffolds, electrospun materials, hydrogels, and particles. The journey finishes with a look at the roadmap of next-generation recombinant silks, especially the development pipeline of this new industry for clinical use.
Collapse
Affiliation(s)
- Chris Holland
- Department of Materials Science and Engineering The University of Sheffield Sir Robert Hadfield Building, Mappin Street Sheffield South Yorkshire S1 3JD UK
| | - Keiji Numata
- Biomacromolecules Research Team RIKEN Center for Sustainable Resource Science 2‐1 Hirosawa Wako Saitama 351‐0198 Japan
| | - Jelena Rnjak‐Kovacina
- Graduate School of Biomedical Engineering The University of New South Wales Sydney NSW 2052 Australia
| | - F. Philipp Seib
- Leibniz Institute of Polymer Research Dresden Max Bergmann Center of Biomaterials Dresden Dresden 01069 Germany
- Strathclyde Institute of Pharmacy and Biomedical Sciences University of Strathclyde Glasgow G4 0RE UK
| |
Collapse
|
35
|
Aghaei-Ghareh-Bolagh B, Guan J, Wang Y, Martin AD, Dawson R, Mithieux SM, Weiss AS. Optically robust, highly permeable and elastic protein films that support dual cornea cell types. Biomaterials 2019; 188:50-62. [DOI: 10.1016/j.biomaterials.2018.10.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/26/2018] [Accepted: 10/07/2018] [Indexed: 10/28/2022]
|
36
|
Deardorff PM, McKay TB, Wang S, Ghezzi CE, Cairns DM, Abbott RD, Funderburgh JL, Kenyon KR, Kaplan DL. Modeling Diabetic Corneal Neuropathy in a 3D In Vitro Cornea System. Sci Rep 2018; 8:17294. [PMID: 30470798 PMCID: PMC6251923 DOI: 10.1038/s41598-018-35917-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 10/31/2018] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus is a disease caused by innate or acquired insulin deficiency, resulting in altered glucose metabolism and high blood glucose levels. Chronic hyperglycemia is linked to development of several ocular pathologies affecting the anterior segment, including diabetic corneal neuropathy and keratopathy, neovascular glaucoma, edema, and cataracts leading to significant visual defects. Due to increasing disease prevalence, related medical care costs, and visual impairment resulting from diabetes, a need has arisen to devise alternative systems to study molecular mechanisms involved in disease onset and progression. In our current study, we applied a novel 3D in vitro model of the human cornea comprising of epithelial, stromal, and neuronal components cultured in silk scaffolds to study the pathological effects of hyperglycemia on development of diabetic corneal neuropathy. Specifically, exposure to sustained levels of high glucose, ranging from 35 mM to 45 mM, were applied to determine concentration-dependent effects on nerve morphology, length and density of axons, and expression of metabolic enzymes involved in glucose metabolism. By comparing these metrics to in vivo studies, we have developed a functional 3D in vitro model for diabetic corneal neuropathy as a means to investigate corneal pathophysiology resulting from prolonged exposure to hyperglycemia.
Collapse
Affiliation(s)
- Phillip M Deardorff
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Tina B McKay
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Siran Wang
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Chiara E Ghezzi
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Dana M Cairns
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Rosalyn D Abbott
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - James L Funderburgh
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Kenneth R Kenyon
- Department of Ophthalmology, Tufts New England Medical Center, Boston, MA, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA.
| |
Collapse
|
37
|
Tran SH, Wilson CG, Seib FP. A Review of the Emerging Role of Silk for the Treatment of the Eye. Pharm Res 2018; 35:248. [PMID: 30397820 PMCID: PMC6223815 DOI: 10.1007/s11095-018-2534-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 10/23/2018] [Indexed: 12/12/2022]
Abstract
Silk is a remarkable biopolymer with a long history of medical use. Silk fabrications have a robust track record for load-bearing applications, including surgical threads and meshes, which are clinically approved for use in humans. The progression of top-down and bottom-up engineering approaches using silk as the basis of a drug delivery or cell-loaded matrix helped to re-ignite interest in this ancient material. This review comprehensively summarises the current applications of silk for tissue engineering and drug delivery, with specific reference to the eye. Additionally, the review also covers emerging trends for the use of silk as a biologically active biopolymer for the treatment of eye disorders. The review concludes with future capabilities of silk to contribute to advanced, electronically-enhanced ocular drug delivery concepts.
Collapse
Affiliation(s)
- Simon H Tran
- 37D Biosystems, Inc., 2372 Morse Avenue, Suite 433, Irvine, California, 92614, USA
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - Clive G Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - F Philipp Seib
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK.
- Max Bergmann Center of Biomaterials Dresden, Leibniz Institute of Polymer Research Dresden, Hohe Strasse 6, 01069, Dresden, Germany.
| |
Collapse
|
38
|
Siran W, Ghezzi CE, Cairns DM, Pollard RE, Chen Y, Gomes R, McKay TB, Pouli D, Jamali A, Georgakoudi I, Funderburgh JL, Kenyon K, Hamrah P, Kaplan DL. Human Corneal Tissue Model for Nociceptive Assessments. Adv Healthc Mater 2018; 7:e1800488. [PMID: 30091220 DOI: 10.1002/adhm.201800488] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 06/28/2018] [Indexed: 12/13/2022]
Abstract
New in vitro tissue models to mimic in vivo conditions are needed to provide insight into mechanisms involved in peripheral pain responses, potential therapeutic strategies to address these responses, and to replace animal models for such indications. For example, the rabbit cornea Draize test has become the standard method used for decades to screen ophthalmic drug and consumer product toxicity. In vitro tissue models with functional innervation have the potential to replace in vivo animal testing and provide sophisticated bench tools to study ocular nociception and its amelioration. Herein, full thickness, innervated, 3D human corneal tissues are grown under physiologically relevant culture conditions to study nociceptive-related responses, by mimicking ocular environmental cues, including intraocular pressure (IOP) and tear flow (TF). Capsaicin, a chili pepper-derived irritant known to cause a burning sensation in mammalian tissues is utilized as a nociceptive stimulant to induce pain, while subsequent serum treatment is used to mimic healing. Pain mediators released upon capsaicin stimulation and cell regrowth after serum treatment are characterized to assess ocular responses in this new, innervated, human corneal tissue system for comparison of outcomes to established animal and related responses.
Collapse
Affiliation(s)
- Wang Siran
- Department of Biomedical EngineeringTufts University 4 Colby Street Medford MA 02155 USA
| | - Chiara E. Ghezzi
- Department of Biomedical EngineeringTufts University 4 Colby Street Medford MA 02155 USA
| | - Dana M. Cairns
- Department of Biomedical EngineeringTufts University 4 Colby Street Medford MA 02155 USA
| | - Rachel E. Pollard
- Department of Biomedical EngineeringTufts University 4 Colby Street Medford MA 02155 USA
| | - Ying Chen
- Department of Biomedical EngineeringTufts University 4 Colby Street Medford MA 02155 USA
| | - Rachel Gomes
- Department of Biomedical EngineeringTufts University 4 Colby Street Medford MA 02155 USA
- School of MedicineDepartamento de Oftalmologia da Escola Paulista de MedicinaFederal University of São Paulo Botucatu, 822 – Vila Clementino São Paulo –SP 04023‐062 Brazil
| | - Tina B. McKay
- Department of Biomedical EngineeringTufts University 4 Colby Street Medford MA 02155 USA
| | - Dimitra Pouli
- Department of Biomedical EngineeringTufts University 4 Colby Street Medford MA 02155 USA
| | - Arsia Jamali
- New England Eye CenterTufts Medical Center 260 Tremont St, 9th Floor Boston MA 02111 USA
| | - Irene Georgakoudi
- Department of Biomedical EngineeringTufts University 4 Colby Street Medford MA 02155 USA
| | - James L. Funderburgh
- Eye & Ear InstituteDepartment of OphthalmologyUniversity of Pittsburgh 203 Lothrop Street, Room 1011 Pittsburgh PA 15213 USA
| | - Kenneth Kenyon
- New England Eye CenterTufts Medical Center 260 Tremont St, 9th Floor Boston MA 02111 USA
| | - Pedram Hamrah
- New England Eye CenterTufts Medical Center 260 Tremont St, 9th Floor Boston MA 02111 USA
| | - David L. Kaplan
- Department of Biomedical EngineeringTufts University 4 Colby Street Medford MA 02155 USA
| |
Collapse
|
39
|
Abbott A, Oxburgh L, Kaplan DL, Coburn JM. Avidin Adsorption to Silk Fibroin Films as a Facile Method for Functionalization. Biomacromolecules 2018; 19:3705-3713. [PMID: 30041518 DOI: 10.1021/acs.biomac.8b00824] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Silk fibroin biomaterials are highly versatile in terms of materials formation and functionalization, with applications in tissue engineering and drug delivery, but necessitate modifications for optimized biological activity. Herein, a facile, avidin-based technique is developed to noncovalently functionalize silk materials with bioactive molecules. The ability to adsorb avidin to silk surfaces and subsequently couple biotinylated macromolecules via avidin-biotin interaction is described. This method better preserved functionality than standard covalent coupling techniques using carbodiimide cross-linking chemistry. The controlled release of avidin from the silk surface was demonstrated by altering the adsorption parameters. Application of this technique to culturing human foreskin fibroblasts (hFFs) and human mesenchymal stem cells (hMSCs) on arginine-glycine-aspartic-acid-modified (RGD-modified) silk showed increased cell growth over a seven-day period. This technique provides a facile method for the versatile functionalization of silk materials for biomedical applications including tissue engineering, drug delivery, and biological sensing.
Collapse
Affiliation(s)
- Alycia Abbott
- Worcester Polytechnic Institute , Worcester , Massachusetts 01605 , United States
| | - Leif Oxburgh
- Maine Medical Center Research Institute , Scarborough , Maine 04074 , United States
| | - David L Kaplan
- Tufts University , Medford , Massachusetts 02155 , United States
| | - Jeannine M Coburn
- Worcester Polytechnic Institute , Worcester , Massachusetts 01605 , United States.,Tufts University , Medford , Massachusetts 02155 , United States
| |
Collapse
|
40
|
Palchesko RN, Carrasquilla SD, Feinberg AW. Natural Biomaterials for Corneal Tissue Engineering, Repair, and Regeneration. Adv Healthc Mater 2018; 7:e1701434. [PMID: 29845780 DOI: 10.1002/adhm.201701434] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 03/01/2018] [Indexed: 12/13/2022]
Abstract
Corneal blindness is a major cause of vision loss, estimated to affect over 10 million people worldwide. Once impaired through clouding or shape change, the best treatment option for restoring vision is corneal transplantation using full or partial thickness cadaveric grafts. However, donor corneas are globally limited and face rejection and graft failure, similar to other transplanted organs. Thus, there is a need for viable alternatives to donor corneas in order to increase supply, reduce rejection, and to minimize variability in tissue quality. To address this, researchers have developed new materials and strategies to tissue engineer full or partial thickness cornea grafts in order to repair, regenerate, or replace the diseased cornea. This progress report first reviews the anatomy and physiology of the cornea to frame the biological requirements and discuss the injuries and diseases that necessitate the need fortransplantation, as well as the requirements for a suitable donor tissue alternative. This is followed by recent progress using naturally derived biomaterials including silk, collagen, amniotic membranes, and decellularized corneas. Finally, remaining challenges in the field as they relate to the biomaterials discussed are identified, and the future research directions that should result in further advances in restoring corneal vision are highlighted.
Collapse
Affiliation(s)
- Rachelle N. Palchesko
- Department of Biomedical Engineering; Carnegie Mellon University; Pittsburgh PA 15213 USA
- Louis J. Fox Center for Vision Restoration; University of Pittsburgh and UPMC; Pittsburgh PA 15213 USA
| | | | - Adam W. Feinberg
- Department of Biomedical Engineering; Carnegie Mellon University; Pittsburgh PA 15213 USA
- Louis J. Fox Center for Vision Restoration; University of Pittsburgh and UPMC; Pittsburgh PA 15213 USA
- Department of Materials Science and Engineering; Carnegie Mellon University; Pittsburgh PA 15213 USA
| |
Collapse
|
41
|
Prina E, Mistry P, Sidney LE, Yang J, Wildman RD, Bertolin M, Breda C, Ferrari B, Barbaro V, Hopkinson A, Dua HS, Ferrari S, Rose FRAJ. 3D Microfabricated Scaffolds and Microfluidic Devices for Ocular Surface Replacement: a Review. Stem Cell Rev Rep 2018; 13:430-441. [PMID: 28573367 DOI: 10.1007/s12015-017-9740-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent years, there has been increased research interest in generating corneal substitutes, either for use in the clinic or as in vitro corneal models. The advancement of 3D microfabrication technologies has allowed the reconstruction of the native microarchitecture that controls epithelial cell adhesion, migration and differentiation. In addition, such technology has allowed the inclusion of a dynamic fluid flow that better mimics the physiology of the native cornea. We review the latest innovative products in development in this field, from 3D microfabricated hydrogels to microfluidic devices.
Collapse
Affiliation(s)
- Elisabetta Prina
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Pritesh Mistry
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Laura E Sidney
- Academic Ophthalmology, Division of Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Jing Yang
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Ricky D Wildman
- Faculty of Engineering, University of Nottingham, Nottingham, UK
| | - Marina Bertolin
- Fondazione Banca degli Occhi del Veneto, c/o Padiglione G. Rama - Via Paccagnella 11, 30174 Zelarino, Venice, Italy
| | - Claudia Breda
- Fondazione Banca degli Occhi del Veneto, c/o Padiglione G. Rama - Via Paccagnella 11, 30174 Zelarino, Venice, Italy
| | - Barbara Ferrari
- Fondazione Banca degli Occhi del Veneto, c/o Padiglione G. Rama - Via Paccagnella 11, 30174 Zelarino, Venice, Italy
| | - Vanessa Barbaro
- Fondazione Banca degli Occhi del Veneto, c/o Padiglione G. Rama - Via Paccagnella 11, 30174 Zelarino, Venice, Italy
| | - Andrew Hopkinson
- Academic Ophthalmology, Division of Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Harminder S Dua
- Academic Ophthalmology, Division of Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Stefano Ferrari
- Fondazione Banca degli Occhi del Veneto, c/o Padiglione G. Rama - Via Paccagnella 11, 30174 Zelarino, Venice, Italy.
| | - Felicity R A J Rose
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| |
Collapse
|
42
|
Matthyssen S, Van den Bogerd B, Dhubhghaill SN, Koppen C, Zakaria N. Corneal regeneration: A review of stromal replacements. Acta Biomater 2018; 69:31-41. [PMID: 29374600 DOI: 10.1016/j.actbio.2018.01.023] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 01/16/2018] [Accepted: 01/16/2018] [Indexed: 12/13/2022]
Abstract
Corneal blindness is traditionally treated by transplantation of a donor cornea, or in severe cases by implantation of an artificial cornea or keratoprosthesis. Due to severe donor shortages and the risks of complications that come with artificial corneas, tissue engineering in ophthalmology has become more focused on regenerative strategies using biocompatible materials either with or without cells. The stroma makes up the bulk of the corneal thickness and mainly consists of a tightly interwoven network of collagen type I, making it notoriously difficult to recreate in a laboratory setting. Despite the challenges that come with corneal stromal tissue engineering, there has recently been enormous progress in this field. A large number of research groups are working towards developing the ideal biomimetic, cytocompatible and transplantable stromal replacement. Here we provide an overview of the approaches directed towards tissue engineering the corneal stroma, from classical collagen gels, films and sponges to less traditional components such as silk, fish scales, gelatin and polymers. The perfect stromal replacement has yet to be identified and future research should be directed at combined approaches, in order to not only host native stromal cells but also restore functionality. STATEMENT OF SIGNIFICANCE In the field of tissue engineering and regenerative medicine in ophthalmology the focus has shifted towards a common goal: to restore the corneal stroma and thereby provide a new treatment option for patients who are currently blind due to corneal opacification. Currently the waiting lists for corneal transplantation include more than 10 million patients, due to severe donor shortages. Alternatives to the transplantation of a donor cornea include the use of artificial cornea, but these are by no means biomimetic and therefore do not provide good outcomes. In recent years a lot of work has gone into the development of tissue engineered scaffolds and other biomaterials suitable to replace the native stromal tissue. Looking at all the different approaches separately is a daunting task and up until now there was no review article in which every approach is discussed. This review does include all approaches, from classical tissue engineering with collagen to the use of various alternative biomaterials and even fish scales. Therefore, this review can serve as a reference work for those starting in the field and but also to stimulate collaborative efforts in the future.
Collapse
|
43
|
Chen J, Zhang W, Kelk P, Backman LJ, Danielson P. Substance P and patterned silk biomaterial stimulate periodontal ligament stem cells to form corneal stroma in a bioengineered three-dimensional model. Stem Cell Res Ther 2017; 8:260. [PMID: 29132420 PMCID: PMC5683543 DOI: 10.1186/s13287-017-0715-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/16/2017] [Accepted: 10/26/2017] [Indexed: 12/13/2022] Open
Abstract
Background We aimed to generate a bioengineered multi-lamellar human corneal stroma tissue in vitro by differentiating periodontal ligament stem cells (PDLSCs) towards keratocytes on an aligned silk membrane. Methods Human PDLSCs were isolated and identified. The neuropeptide substance P (SP) was added in keratocyte differentiation medium (KDM) to evaluate its effect on keratocyte differentiation of PDLSCs. PDLSCs were then seeded on patterned silk membrane and cultured with KDM and SP. Cell alignment was evaluated and the expression of extracellular matrix (ECM) components of corneal stroma was detected. Finally, multi-lamellar tissue was constructed in vitro by PDLSCs seeded on patterned silk membranes, which were stacked orthogonally and stimulated by KDM supplemented with SP for 18 days. Sections were prepared and subsequently stained with hematoxylin and eosin or antibodies for immunofluorescence observation of human corneal stroma-related proteins. Results SP promoted the expression of corneal stroma-related collagens (collagen types I, III, V, and VI) during the differentiation induced by KDM. Patterned silk membrane guided cell alignment of PDLSCs, and important ECM components of the corneal stroma were shown to be deposited by the cells. The constructed multi-lamellar tissue was found to support cells growing between every two layers and expressing the main type of collagens (collagen types I and V) and proteoglycans (lumican and keratocan) of normal human corneal stroma. Conclusions Multi-lamellar human corneal stroma-like tissue can be constructed successfully in vitro by PDLSCs seeded on orthogonally aligned, multi-layered silk membranes with SP supplementation, which shows potential for future corneal tissue engineering. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0715-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jialin Chen
- Department of Integrative Medical Biology, Anatomy, Umeå University, SE-901 87, Umeå, Sweden
| | - Wei Zhang
- Department of Integrative Medical Biology, Anatomy, Umeå University, SE-901 87, Umeå, Sweden
| | - Peyman Kelk
- Department of Integrative Medical Biology, Anatomy, Umeå University, SE-901 87, Umeå, Sweden
| | - Ludvig J Backman
- Department of Integrative Medical Biology, Anatomy, Umeå University, SE-901 87, Umeå, Sweden.,Department of Community Medicine and Rehabilitation, Physiotherapy, Umeå University, Umeå, Sweden
| | - Patrik Danielson
- Department of Integrative Medical Biology, Anatomy, Umeå University, SE-901 87, Umeå, Sweden. .,Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden.
| |
Collapse
|
44
|
Gouveia RM, Koudouna E, Jester J, Figueiredo F, Connon CJ. Template Curvature Influences Cell Alignment to Create Improved Human Corneal Tissue Equivalents. ACTA ACUST UNITED AC 2017; 1:e1700135. [DOI: 10.1002/adbi.201700135] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 08/30/2017] [Indexed: 12/28/2022]
Affiliation(s)
- Ricardo M. Gouveia
- Institute of Genetic Medicine; Newcastle University; Newcastle upon Tyne NE1 3BZ UK
| | - Elena Koudouna
- Gavin Herbert Eye Institute; University of California Irvine; Irvine CA 92697 USA
- Structural Biophysics Research Group; School of Optometry and Vision Sciences; Cardiff University; Cardiff CF24 4HQ Wales UK
| | - James Jester
- Gavin Herbert Eye Institute; University of California Irvine; Irvine CA 92697 USA
| | - Francisco Figueiredo
- Institute of Genetic Medicine; Newcastle University; Newcastle upon Tyne NE1 3BZ UK
- Department of Ophthalmology; Royal Victoria Infirmary; Newcastle upon Tyne NE1 4LP UK
| | - Che J. Connon
- Institute of Genetic Medicine; Newcastle University; Newcastle upon Tyne NE1 3BZ UK
| |
Collapse
|
45
|
Gosselin EA, Torregrosa T, Ghezzi CE, Mendelsohn AC, Gomes R, Funderburgh JL, Kaplan DL. Multi-layered silk film coculture system for human corneal epithelial and stromal stem cells. J Tissue Eng Regen Med 2017; 12:285-295. [PMID: 28600807 DOI: 10.1002/term.2499] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 03/15/2017] [Accepted: 06/07/2017] [Indexed: 01/22/2023]
Abstract
With insufficient options to meet the clinical demand for cornea transplants, one emerging area of emphasis is on cornea tissue engineering. In the present study, the goal was to combine the corneal stroma and epithelium into one coculture system, to monitor both human corneal stromal stem cell (hCSSC) and human corneal epithelial cell (hCE) growth and differentiation into keratocytes and differentiated epithelium in these three-dimensional tissue systems in vitro. Coculture conditions were first optimized, including the medium, air-liquid interface culture, and surface topography and chemistry of biomaterial scaffold films based on silk protein. The silk was used as scaffolding for both stromal and epithelial tissue layers because it is cell compatible, can be surface patterned, and is optically clear. Next, the effects of proliferating and differentiating hCEs and hCSSCs were studied in this in vitro system, including the effects on cell proliferation, matrix formation by immunochemistry, and gene expression by quantitative reverse transcription-polymerase chain reaction. The incorporation of both cell types into the coculture system demonstrated more complete differentiation and growth for both cell types compared to the corneal stromal cells and corneal epithelial cells alone. Silk films for corneal epithelial culture were optimized to combine a 4.0-μm-scale surface pattern with bulk-loaded collagen type IV. Differentiation of each cell type was in evidence based on increased expression of corneal stroma and epithelial proteins and transcript levels after 6 weeks in coculture on the optimized silk scaffolds.
Collapse
Affiliation(s)
- Emily A Gosselin
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Tess Torregrosa
- Department of Chemical Engineering, Tufts University, Medford, MA, USA
| | - Chiara E Ghezzi
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | | | - Rachel Gomes
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - James L Funderburgh
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| |
Collapse
|
46
|
Influence of Micropatterned Silk Fibroin Films on Human Umbilical Endothelial Cell Behaviors. J Med Biol Eng 2017. [DOI: 10.1007/s40846-017-0249-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
47
|
Wound-Healing Studies in Cornea and Skin: Parallels, Differences and Opportunities. Int J Mol Sci 2017; 18:ijms18061257. [PMID: 28604651 PMCID: PMC5486079 DOI: 10.3390/ijms18061257] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 05/24/2017] [Accepted: 05/31/2017] [Indexed: 02/06/2023] Open
Abstract
The cornea and the skin are both organs that provide the outer barrier of the body. Both tissues have developed intrinsic mechanisms that protect the organism from a wide range of external threats, but at the same time also enable rapid restoration of tissue integrity and organ-specific function. The easy accessibility makes the skin an attractive model system to study tissue damage and repair. Findings from skin research have contributed to unravelling novel fundamental principles in regenerative biology and the repair of other epithelial-mesenchymal tissues, such as the cornea. Following barrier disruption, the influx of inflammatory cells, myofibroblast differentiation, extracellular matrix synthesis and scar formation present parallel repair mechanisms in cornea and skin wound healing. Yet, capillary sprouting, while pivotal in proper skin wound healing, is a process that is rather associated with pathological repair of the cornea. Understanding the parallels and differences of the cellular and molecular networks that coordinate the wound healing response in skin and cornea are likely of mutual importance for both organs with regard to the development of regenerative therapies and understanding of the disease pathologies that affect epithelial-mesenchymal interactions. Here, we review the principal events in corneal wound healing and the mechanisms to restore corneal transparency and barrier function. We also refer to skin repair mechanisms and their potential implications for regenerative processes in the cornea.
Collapse
|
48
|
Peterová E, Podmolíková L, Řezáčová M, Mrkvicová A. Fibroblast Growth Factor-1 Suppresses TGF-β-Mediated Myofibroblastic Differentiation of Rat Hepatic Stellate Cells. ACTA MEDICA (HRADEC KRÁLOVÉ) 2017; 59:124-132. [PMID: 28440215 DOI: 10.14712/18059694.2017.39] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Myofibroblast expansion is a critical event in the pathogenesis of liver fibrosis. The activation of hepatic stellate cells (HSC) to myofibroblast (MFB) results in the enhanced production of extracellular matrix (ECM). In this study, we explored the effect of acidic fibroblast growth factor (FGF-1) treatment on a transforming growth factor (TGF-β1) induced MFB conversion. We used HSC-T6 cell line, which represents well-established model of activated HSC. These cells strongly expressed α-smooth muscle actin (α-SMA) and fibronectin (FN-EDA) after stimulation with TGF-β1, which is a stimulus for MFB differentiation and ECM production. FGF-1 reduced proteins expression to levels comparable with untreated cells. Mild repression of secreted gelatinases was seen in culture media after FGF-1 treatment. The exposure of cells to collagen gel leads to changes in cell morphology and in expression of MFB markers. Lack of α-SMA in cells embedded to collagen gel was detected. When stimulated with TGF-β1, the cells increased expression of FN-EDA, but not α-SMA. Although the cells on plastic and in collagen gel show different properties, FGF-1 reduced expression of FN-EDA in both conditions. Disrupting TGF-β1 signalling pathway represents a potential strategy for the treatment of fibrosis. We showed that FGF-1 could antagonize signals initiated by TGF-β1.
Collapse
Affiliation(s)
- Eva Peterová
- Department of Medical Biochemistry, Charles University, Faculty of Medicine in Hradec Králové, Hradec Králové, Czech Republic
| | - Lucie Podmolíková
- Department of Medical Biochemistry, Charles University, Faculty of Medicine in Hradec Králové, Hradec Králové, Czech Republic
| | - Martina Řezáčová
- Department of Medical Biochemistry, Charles University, Faculty of Medicine in Hradec Králové, Hradec Králové, Czech Republic
| | - Alena Mrkvicová
- Department of Medical Biochemistry, Charles University, Faculty of Medicine in Hradec Králové, Hradec Králové, Czech Republic.
| |
Collapse
|
49
|
Syed-Picard FN, Du Y, Hertsenberg AJ, Palchesko R, Funderburgh ML, Feinberg AW, Funderburgh JL. Scaffold-free tissue engineering of functional corneal stromal tissue. J Tissue Eng Regen Med 2017; 12:59-69. [PMID: 27863068 DOI: 10.1002/term.2363] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 09/30/2016] [Accepted: 11/09/2016] [Indexed: 12/13/2022]
Abstract
Blinding corneal scarring is predominately treated with allogeneic graft tissue; however, there is a worldwide shortage of donor tissue leaving millions in need of therapy. Human corneal stromal stem cells (CSSC) have been shown produce corneal tissue when cultured on nanofibre scaffolding, but this tissue cannot be readily separated from the scaffold. In this study, scaffold-free tissue engineering methods were used to generate biomimetic corneal stromal tissue constructs that can be transplanted in vivo without introducing the additional variables associated with exogenous scaffolding. CSSC were cultured on substrates with aligned microgrooves, which directed parallel cell alignment and matrix organization, similar to the organization of native corneal stromal lamella. CSSC produced sufficient matrix to allow manual separation of a tissue sheet from the grooved substrate. These constructs were cellular and collagenous tissue sheets, approximately 4 μm thick and contained extracellular matrix molecules typical of corneal tissue including collagen types I and V and keratocan. Similar to the native corneal stroma, the engineered corneal tissues contained long parallel collagen fibrils with uniform diameter. After being transplanted into mouse corneal stromal pockets, the engineered corneal stromal tissues became transparent, and the human CSSCs continued to express human corneal stromal matrix molecules. Both in vitro and in vivo, these scaffold-free engineered constructs emulated stromal lamellae of native corneal stromal tissues. Scaffold-free engineered corneal stromal constructs represent a novel, potentially autologous, cell-generated, biomaterial with the potential for treating corneal blindness. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
| | - Yiqin Du
- Department of Ophthalmology, University of Pittsburgh, PA, USA.,McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA
| | | | - Rachelle Palchesko
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | - Adam W Feinberg
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA.,Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.,Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - James L Funderburgh
- Department of Ophthalmology, University of Pittsburgh, PA, USA.,McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA
| |
Collapse
|
50
|
|