1
|
Kauppila M, Mörö A, Valle-Delgado JJ, Huhtanen S, Hopia K, Österberg M, Skottman H. COMPARTMENTALIZED 3D BIOPRINTING OF THE LIMBAL NICHE WITH DISTINCT HPSC-LSC SUBPOPULATIONS FOR CORNEAL DISEASE MODELING. Acta Biomater 2025:S1742-7061(25)00404-0. [PMID: 40449707 DOI: 10.1016/j.actbio.2025.05.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 05/16/2025] [Accepted: 05/28/2025] [Indexed: 06/03/2025]
Abstract
Limbal epithelial stem cells (LSCs) are essential for corneal epithelium regeneration and visual acuity. The limbal niche's physicochemical properties regulate LSC function, but their role is not fully understood. Developing in vitro models that mimic the native niche can enhance our understanding of niche functions, despite the challenges of niche complexity. In this study, we created a 3D bioprinted limbal niche model using a hybrid approach that combines two human pluripotent stem cell-derived LSC (hPSC-LSC) subpopulations (p63+ and ABCG2+ cells) within hyaluronic acid (HA)-based bioinks and a stiff polyacrylamide (PA) gel scaffold produced by conventional gel casting. We analyzed the mechanical properties of the bioinks and assessed cell viability, morphology, and protein expression after one week of culture. Finally, we conducted a proof-of-concept wound healing assay using an alkali burn injury model to assess the functionality of the model for research purposes. The results show that this 3D model effectively replicated the mechanical environment of native tissue, maintains stability for one-week post-printing, and supports LSC viability and normal in vitro phenotype. In addition, the wound healing assay showed a cellular response, indicated by non-simultaneous caspase-3 activation of hPSC-LSC subpopulations for 48 hours post-wounding. This model provides a valuable platform for investigating the limbal niche and advancing cellular therapies applicable to other tissue niches throughout the body. STATEMENT OF SIGNIFICANCE: The corneal limbal niche is crucial for corneal regeneration, creating a high demand for in vitro models. However, current models are not sufficiently replicating the complexity of native tissue and importantly, lack the element of recently demostrated limbal stem cell (LSC) heterogeneity. In this study, we combine three key features of the limbus, including stiffness, architecture and compartmentalization, to create limbal niche-mimicking structures using 3D bioprinting with two human pluripotent stem cell derived LSC (hPSC-LSC) subpopulations. We demonstrate structural stability, native tissue-like mechanical properties, sustained cellular viability, stable hPSC-LSC phenotype post-printing, and a tissue-mimicking response to wounding. This approach offers an innovative strategy to model complex niches and advance the understanding of limbal niche functions.
Collapse
Affiliation(s)
- M Kauppila
- Eye Regeneration Group, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
| | - A Mörö
- Eye Regeneration Group, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
| | - J J Valle-Delgado
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, Espoo, 02150 Finland
| | - S Huhtanen
- Eye Regeneration Group, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
| | - K Hopia
- Eye Regeneration Group, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
| | - M Österberg
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, Espoo, 02150 Finland
| | - H Skottman
- Eye Regeneration Group, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland.
| |
Collapse
|
2
|
Islam MM, Saha A, Trisha FA, Gonzalez-Andrades M, Patra HK, Griffith M, Chodosh J, Rajaiya J. An in vitro 3-dimensional Collagen-based Corneal Construct with Innervation Using Human Corneal Cell Lines. OPHTHALMOLOGY SCIENCE 2024; 4:100544. [PMID: 39139547 PMCID: PMC11321308 DOI: 10.1016/j.xops.2024.100544] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 08/15/2024]
Abstract
Purpose To develop a 3-dimensional corneal construct suitable for in vitro studies of disease conditions and therapies. Design In vitro human corneal constructs were created using chemically crosslinked collagen and chondroitin sulfate extracellular matrix and seeded with 3 human corneal cell types (epithelial, stromal, and endothelial) together with neural cells. The neural cells were derived from hybrid neuroblastoma cells and the other cells used from immortalized human corneal cell lines. To check the feasibility and characterize the constructs, cytotoxicity, cell proliferation, histology, and protein expression studies were performed. Results Optimized culture condition permitted synchronized viability across the cell types within the construct. The construct showed a typical appearance for different cellular layers, including healthy appearing, phenotypically differentiated neurons. The expected protein expression profiles for specific cell types within the construct were confirmed with western blotting. Conclusions An in vitro corneal construct was successfully developed with maintenance of individual cell phenotypes with anatomically correct cellular loci. The construct may be useful in evaluation of specific corneal disorders and in developing different corneal disease models. Additionally, the construct can be used in evaluating drug targeting and/or penetration to individual corneal layers, testing novel therapeutics for corneal diseases, and potentially reducing the necessity for animals in corneal research at the early stages of investigation. Financial Disclosures Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Mohammad Mirazul Islam
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear and Schepens Eye Research Institute, Boston, Massachusetts
- Department of Ophthalmology and Visual Sciences, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Amrita Saha
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear and Schepens Eye Research Institute, Boston, Massachusetts
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Farzana Afrose Trisha
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear and Schepens Eye Research Institute, Boston, Massachusetts
| | - Miguel Gonzalez-Andrades
- Department of Ophthalmology, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital and University of Cordoba, Cordoba, Spain
| | - Hirak K. Patra
- UCL Division of Surgery and Interventional Science, Department of Surgical Biotechnology, University College London, London, UK
| | - May Griffith
- Department of Ophthalmology, Université de Montréal and Centre de recherche de l'Hôpital Maisonneuve Rosemont, Montreal, Quebec, Canada
| | - James Chodosh
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear and Schepens Eye Research Institute, Boston, Massachusetts
- Department of Ophthalmology and Visual Sciences, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Jaya Rajaiya
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear and Schepens Eye Research Institute, Boston, Massachusetts
- Department of Ophthalmology and Visual Sciences, University of New Mexico School of Medicine, Albuquerque, New Mexico
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, New Mexico
| |
Collapse
|
3
|
Mantry S, Behera A, Pradhan S, Mohanty L, Kumari R, Singh A, Yadav MK. Polysaccharide-based chondroitin sulfate macromolecule loaded hydrogel/scaffolds in wound healing- A comprehensive review on possibilities, research gaps, and safety assessment. Int J Biol Macromol 2024; 279:135410. [PMID: 39245102 DOI: 10.1016/j.ijbiomac.2024.135410] [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: 06/14/2024] [Revised: 08/20/2024] [Accepted: 09/05/2024] [Indexed: 09/10/2024]
Abstract
Wound healing is an intricate multifactorial process that may alter the extent of scarring left by the wound. A substantial portion of the global population is impacted by non-healing wounds, imposing significant financial burdens on the healthcare system. The conventional dosage forms fail to improve the condition, especially in the presence of other morbidities. Thus, there is a pressing requirement for a type of wound dressing that can safeguard the wound site and facilitate skin regeneration, ultimately expediting the healing process. In this context, Chondroitin sulfate (CS), a sulfated glycosaminoglycan material, is capable of hydrating tissues and further promoting the healing. Thus, this comprehensive review article delves into the recent advancement of CS-based hydrogel/scaffolds for wound healing management. The article initially summarizes the various physicochemical characteristics and sources of CS, followed by a brief understanding of the importance of hydrogel and CS in tissue regeneration processes. This is the first instance of such a comprehensive summarization of CS-based hydrogel/scaffolds in wound healing, focusing more on the mechanistic wound healing process, furnishing the recent innovations and toxicity profile. This contemporary review provides a profound acquaintance of strategies for contemporary challenges and future direction in CS-based hydrogel/scaffolds for wound healing.
Collapse
Affiliation(s)
- Shubhrajit Mantry
- Department of Pharmaceutics, Department of Pharmacy, Sarala Birla University, Birla Knowledge City, Ranchi 835103, Jharkhand, India.
| | - Ashutosh Behera
- Department of Pharmaceutical Quality Assurance, Department of Pharmacy, Sarala Birla University, Birla Knowledge City, Ranchi 835103, Jharkhand, India; Department of Pharmaceutical Quality Assurance, Florence College of Pharmacy, IRBA, Ranchi, 835103, Jharkhand, India
| | - Shaktiprasad Pradhan
- Department of Pharmaceutical Chemistry, Koustuv Research Institute of Medical Science (KRIMS), Koustuv Technical Campus, Patia, Bhubaneswar, Odisha 751024, India
| | - Lalatendu Mohanty
- Department of Pharmacology, Department of Pharmaceutical Sciences, HNB Garhwal University (A Central University), Tehri Garhwal, Uttarakhand 24916, India
| | - Ragni Kumari
- School of Pharmacy, LNCT University, Bhopal 462022, Madhya Pradesh, India
| | - Ankita Singh
- Department of Pharmacy, Faculty of Medical Science & Research (FMSR), Sai Nath University, Ranchi, Jharkhand 835219, India
| | - Mahesh Kumar Yadav
- Department of Pharmacy, Faculty of Medical Science & Research (FMSR), Sai Nath University, Ranchi, Jharkhand 835219, India
| |
Collapse
|
4
|
Kumar R, Sinha NR, Mohan RR. Corneal gene therapy: Structural and mechanistic understanding. Ocul Surf 2023; 29:279-297. [PMID: 37244594 PMCID: PMC11926995 DOI: 10.1016/j.jtos.2023.05.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023]
Abstract
Cornea, a dome-shaped and transparent front part of the eye, affords 2/3rd refraction and barrier functions. Globally, corneal diseases are the leading cause of vision impairment. Loss of corneal function including opacification involve the complex crosstalk and perturbation between a variety of cytokines, chemokines and growth factors generated by corneal keratocytes, epithelial cells, lacrimal tissues, nerves, and immune cells. Conventional small-molecule drugs can treat mild-to-moderate traumatic corneal pathology but requires frequent application and often fails to treat severe pathologies. The corneal transplant surgery is a standard of care to restore vision in patients. However, declining availability and rising demand of donor corneas are major concerns to maintain ophthalmic care. Thus, the development of efficient and safe nonsurgical methods to cure corneal disorders and restore vision in vivo is highly desired. Gene-based therapy has huge potential to cure corneal blindness. To achieve a nonimmunogenic, safe and sustained therapeutic response, the selection of a relevant genes, gene editing methods and suitable delivery vectors are vital. This article describes corneal structural and functional features, mechanistic understanding of gene therapy vectors, gene editing methods, gene delivery tools, and status of gene therapy for treating corneal disorders, diseases, and genetic dystrophies.
Collapse
Affiliation(s)
- Rajnish Kumar
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, 65201, USA; One-health One-medicine Vision Research Program, Departments of Veterinary Medicine and Surgery & Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65211, USA; Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow campus, UP, 226028, India
| | - Nishant R Sinha
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, 65201, USA; One-health One-medicine Vision Research Program, Departments of Veterinary Medicine and Surgery & Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65211, USA
| | - Rajiv R Mohan
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, 65201, USA; One-health One-medicine Vision Research Program, Departments of Veterinary Medicine and Surgery & Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65211, USA; Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO, 65212, USA.
| |
Collapse
|
5
|
Sinha NR, Tripathi R, Balne PK, Suleiman L, Simkins K, Chaurasia SS, Mohan RR. Mustard Gas Exposure Actuates SMAD2/3 Signaling to Promote Myofibroblast Generation in the Cornea. Cells 2023; 12:1533. [PMID: 37296653 PMCID: PMC10252656 DOI: 10.3390/cells12111533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Sulfur mustard gas (SM) is a vesicating and alkylating agent used as a chemical weapon in many mass-casualty incidents since World War I. Ocular injuries were reported in >90% of exposed victims. The mechanisms underlying SM-induced blindness remain elusive. This study tested the hypothesis that SM-induced corneal fibrosis occurs due to the generation of myofibroblasts from resident fibroblasts via the SMAD2/3 signaling pathway in rabbit eyes in vivo and primary human corneal fibroblasts (hCSFs) isolated from donor corneas in vitro. Fifty-four New Zealand White Rabbits were divided into three groups (Naïve, Vehicle, SM-Vapor treated). The SM-Vapor group was exposed to SM at 200 mg-min/m3 for 8 min at the MRI Global facility. Rabbit corneas were collected on day 3, day 7, and day 14 for immunohistochemistry, RNA, and protein lysates. SM caused a significant increase in SMAD2/3, pSMAD, and ɑSMA expression on day 3, day 7, and day 14 in rabbit corneas. For mechanistic studies, hCSFs were treated with nitrogen mustard (NM) or NM + SIS3 (SMAD3-specific inhibitor) and collected at 30 m, 8 h, 24 h, 48 h, and 72 h. NM significantly increased TGFβ, pSMAD3, and SMAD2/3 levels. On the contrary, inhibition of SMAD2/3 signaling by SIS3 treatment significantly reduced SMAD2/3, pSMAD3, and ɑSMA expression in hCSFs. We conclude that SMAD2/3 signaling appears to play a vital role in myofibroblast formation in the cornea following mustard gas exposure.
Collapse
Affiliation(s)
- Nishant R. Sinha
- Departments of Veterinary Medicine & Surgery and Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| | - Ratnakar Tripathi
- Departments of Veterinary Medicine & Surgery and Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| | - Praveen K. Balne
- Departments of Veterinary Medicine & Surgery and Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| | - Laila Suleiman
- Departments of Veterinary Medicine & Surgery and Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Katherine Simkins
- Departments of Veterinary Medicine & Surgery and Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Shyam S. Chaurasia
- Departments of Veterinary Medicine & Surgery and Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA
- Ocular Immunology and Angiogenesis Lab, Department of Ophthalmology & Visual Sciences, Froedtert & Medical College of Wisconsin Eye Institute, Milwaukee, WI 53226, USA
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Rajiv R. Mohan
- Departments of Veterinary Medicine & Surgery and Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
6
|
Development of In Vitro Dry Eye Models to Study Proliferative and Anti-Inflammatory Effects of Allogeneic Serum Eye Drops. Int J Mol Sci 2023; 24:ijms24021567. [PMID: 36675083 PMCID: PMC9864688 DOI: 10.3390/ijms24021567] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/08/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
This study aimed to develop valid in vitro models for preclinical evaluation of proliferative and anti-inflammatory effects of human allogeneic serum eye drops for dry eye disease (DED) treatment. A DED wound healing model was developed by analyzing the influence of coating and serum concentrations on human corneal epithelial (HCE-T) wound closure. Further, intralaboratory variance, freeze-thaw cycle effects, donor variability and stability assays were conducted. Interleukin-1β (IL-1β) and tumor necrosis factor α (TNFα) were used to induce the gene expression of matrix metalloproteinase 9 (MMP9), cyclooxygenase 2 (COX2), transforming growth factor-β (TGFβ) and IL-1β. MMP9 induction was optimized using a design-of-experiments (DoE) approach and applied to examine serum under static and dynamic conditions. MMP9 protein expression was analyzed by ELISA. The DED wound healing model detected proliferative effects of serum down to 1% with a small intralaboratory variance. Serum stability was shown over six months, donor variance could be detected, and freeze-thaw cycle effects did not affect wound closure. Serum decreased MMP9 expression on the gene and protein levels. The induction method was successfully optimized using DoE modeling and transferred to a dynamic setting mimicking tear film fluidics. The DED wound healing and inflammatory DED model present useful in vitro models for the preclinical evaluation of allogeneic serum eye drops without the use of animal experiments.
Collapse
|
7
|
Abstract
Despite rigorous investigations, the hydrogels currently available to replace damaged tissues, such as the cornea, cannot fulfill mechanical and structural requirements and, more importantly, cannot be sutured into host tissues due to the lack of hierarchical structures to dissipate exerted stress. In this report, solution electrospinning of polycaprolactone (PCL), protein-based hydrogel perfusion, and layer-by-layer stacking are used to generate a hydrogel-microfiber composite with varying PCL fiber diameters and hydrogel concentrations. Integrating PCL microfibers into the hydrogel synergistically improves the mechanical properties and suturability of the construct up to 10-fold and 50-fold, respectively, compared to the hydrogel and microfiber scaffolds alone, approaching those of the corneal tissue. Human corneal cells cultured on composites are viable and can spread, proliferate, and retain phenotypic characteristics. Moreover, corneal stromal cells migrate into the scaffold, degrade it, and regenerate the extracellular matrix. The current hydrogel reinforcing system paves the way for producing suturable and, therefore, transplantable tissue constructs with desired mechanical properties.
Collapse
Affiliation(s)
- Sina Sharifi
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, Massachusetts02114, United States
| | - Hannah Sharifi
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, Massachusetts02114, United States
| |
Collapse
|
8
|
Hewitt BJ, Batt JM, Shelton RM, Cooper PR, Landini G, Lucas RA, Wiench MD, Milward MR. A 3D Printed Device for In Vitro Generation of Stratified Epithelia at the Air-Liquid Interface. Tissue Eng Part C Methods 2022; 28:599-609. [PMID: 36047814 DOI: 10.1089/ten.tec.2022.0130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Air-liquid interface (ALI) cultures are used to produce stratified epithelial tissues in vitro, notably for the production of oral mucosal equivalents. Currently, there are few purpose-built devices which aim to enhance the ease and reproducibility of generating such tissue. Most ALI cultures utilise stainless steel grids or cell culture inserts to elevate the matrix or scaffold to the surface of the culture media. Here, a novel buoyant epithelial culture device (BECD) was designed to both contain a fibroblast-seeded collagen hydrogel and float in culture media, thereby automatically maintaining the ALI without further user intervention. BECDs aim to mitigate several issues associated with ALI culture; reducing the chance of media flooding the epithelial layer from physical disturbance, reducing technique-sensitivity for less experienced users, and improving the reproducibility of the epithelia generated. H400 oral squamous cell carcinoma cells cultured in BECDs for 7, 14 and 21 days showed continuous increase in epithelial tissue thickness with expected localisation of epithelial differentiation markers: cytokeratin 5, involucrin and E-cadherin. Fused filament fabrication 3D printing with polypropylene used in BECD production allows for rapid turnover and design iteration, presenting a versatile, adaptable and useful tool for application in in vitro cell culture.
Collapse
Affiliation(s)
- Benjamin John Hewitt
- University of Birmingham, School of Dentistry, 5 Mill Pool Way, Birmingham, B5 7EG, Birmingham, United Kingdom of Great Britain and Northern Ireland, B15 2TT;
| | - Joanna M Batt
- University of Birmingham, School of Dentistry, Birmingham, United Kingdom of Great Britain and Northern Ireland;
| | - Richard Michael Shelton
- University of Birmingham School of Dentistry, Dentistry, Birmingham, Birmingham, United Kingdom of Great Britain and Northern Ireland;
| | - Paul R Cooper
- University of Otago Division of Health Sciences, Dunedin, New Zealand;
| | - Gabriel Landini
- University of Birmingham School of Dentistry, School of Dentistry, Birmingham, Birmingham, United Kingdom of Great Britain and Northern Ireland;
| | - Robert A Lucas
- GlaxoSmithKline Consumer Healthcare, Weybridge, Surrey, United Kingdom of Great Britain and Northern Ireland;
| | - Malgorzata D Wiench
- University of Birmingham School of Dentistry, Dentistry, Birmingham, Birmingham, United Kingdom of Great Britain and Northern Ireland.,University of Birmingham Institute of Cancer and Genomic Sciences, Birmingham, Birmingham, United Kingdom of Great Britain and Northern Ireland;
| | - Michael R Milward
- University of Birmingham School of Dentistry, Dentistry, Birmingham, Birmingham, United Kingdom of Great Britain and Northern Ireland;
| |
Collapse
|
9
|
Islam MM, Chivu A, AbuSamra DB, Saha A, Chowdhuri S, Pramanik B, Dohlman CH, Das D, Argüeso P, Rajaiya J, Patra HK, Chodosh J. Crosslinker-free collagen gelation for corneal regeneration. Sci Rep 2022; 12:9108. [PMID: 35650270 PMCID: PMC9160259 DOI: 10.1038/s41598-022-13146-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/20/2022] [Indexed: 12/13/2022] Open
Abstract
Development of an artificial cornea can potentially fulfil the demand of donor corneas for transplantation as the number of donors is far less than needed to treat corneal blindness. Collagen-based artificial corneas stand out as a regenerative option, having promising clinical outcomes. Collagen crosslinked with chemical crosslinkers which modify the parent functional groups of collagen. However, crosslinkers are usually cytotoxic, so crosslinkers need to be removed from implants completely before application in humans. In addition, crosslinked products are mechanically weak and susceptible to enzymatic degradation. We developed a crosslinker free supramolecular gelation strategy using pyrene conjugated dipeptide amphiphile (PyKC) consisting of lysine and cysteine; in which collagen molecules are intertwined inside the PyKC network without any functional group modification of the collagen. The newly developed collagen implants (Coll-PyKC) are optically transparent and can effectively block UV light, are mechanically and enzymatically stable, and can be sutured. The Coll-PyKC implants support the growth and function of all corneal cells, trigger anti-inflammatory differentiation while suppressing the pro-inflammatory differentiation of human monocytes. Coll-PyKC implants can restrict human adenovirus propagation. Therefore, this crosslinker-free strategy can be used for the repair, healing, and regeneration of the cornea, and potentially other damaged organs of the body.
Collapse
Affiliation(s)
- Mohammad Mirazul Islam
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Harvard Medical School, Boston, MA, 02114, USA
| | - Alexandru Chivu
- Department of Surgical Biotechnology, University College London, London, NW3 2PF, UK
| | - Dina B AbuSamra
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Harvard Medical School, Boston, MA, 02114, USA
| | - Amrita Saha
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Harvard Medical School, Boston, MA, 02114, USA
| | - Sumit Chowdhuri
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Bapan Pramanik
- Department of Chemistry, Ben Gurion University of the Negev, Be'er Sheva, Israel
| | - Claes H Dohlman
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Harvard Medical School, Boston, MA, 02114, USA
| | - Debapratim Das
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Pablo Argüeso
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Harvard Medical School, Boston, MA, 02114, USA
| | - Jaya Rajaiya
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Harvard Medical School, Boston, MA, 02114, USA
| | - Hirak K Patra
- Department of Surgical Biotechnology, University College London, London, NW3 2PF, UK.
| | - James Chodosh
- Department of Ophthalmology, Massachusetts Eye and Ear and Schepens Eye Research Institute, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
10
|
Cosert KM, Kim S, Jalilian I, Chang M, Gates BL, Pinkerton KE, Van Winkle LS, Raghunathan VK, Leonard BC, Thomasy SM. Metallic Engineered Nanomaterials and Ocular Toxicity: A Current Perspective. Pharmaceutics 2022; 14:pharmaceutics14050981. [PMID: 35631569 PMCID: PMC9145553 DOI: 10.3390/pharmaceutics14050981] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/06/2022] [Accepted: 04/18/2022] [Indexed: 02/01/2023] Open
Abstract
The ocular surface, comprised of the transparent cornea, conjunctiva, and protective tear film, forms a protective barrier defending deeper structures of the eye from particulate matter and mechanical trauma. This barrier is routinely exposed to a multitude of naturally occurring and engineered nanomaterials (ENM). Metallic ENMs are particularly ubiquitous in commercial products with a high risk of ocular exposure, such as cosmetics and sunscreens. Additionally, there are several therapeutic uses for metallic ENMs owing to their attractive magnetic, antimicrobial, and functionalization properties. The increasing commercial and therapeutic applications of metallic ENMs come with a high risk of ocular exposure with poorly understood consequences to the health of the eye. While the toxicity of metallic ENMs exposure has been rigorously studied in other tissues and organs, further studies are necessary to understand the potential for adverse effects and inform product usage for individuals whose ocular health may be compromised by injury, disease, or surgical intervention. This review provides an update of current literature on the ocular toxicity of metallic ENMs in vitro and in vivo, as well as the risks and benefits of therapeutic applications of metallic ENMs in ophthalmology.
Collapse
Affiliation(s)
- Krista M. Cosert
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (K.M.C.); (S.K.); (I.J.); (M.C.); (B.L.G.); (B.C.L.)
| | - Soohyun Kim
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (K.M.C.); (S.K.); (I.J.); (M.C.); (B.L.G.); (B.C.L.)
| | - Iman Jalilian
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (K.M.C.); (S.K.); (I.J.); (M.C.); (B.L.G.); (B.C.L.)
| | - Maggie Chang
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (K.M.C.); (S.K.); (I.J.); (M.C.); (B.L.G.); (B.C.L.)
| | - Brooke L. Gates
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (K.M.C.); (S.K.); (I.J.); (M.C.); (B.L.G.); (B.C.L.)
| | - Kent E. Pinkerton
- Center for Health and the Environment, University of California Davis, Davis, CA 95616, USA; (K.E.P.); (L.S.V.W.)
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA
| | - Laura S. Van Winkle
- Center for Health and the Environment, University of California Davis, Davis, CA 95616, USA; (K.E.P.); (L.S.V.W.)
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA
| | - Vijay Krishna Raghunathan
- Department of Basic Sciences, College of Optometry, University of Houston, Houston, TX 77004, USA;
- The Ocular Surface Institute, College of Optometry, University of Houston, Houston, TX 77004, USA
- Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, TX 77204, USA
| | - Brian C. Leonard
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (K.M.C.); (S.K.); (I.J.); (M.C.); (B.L.G.); (B.C.L.)
| | - Sara M. Thomasy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (K.M.C.); (S.K.); (I.J.); (M.C.); (B.L.G.); (B.C.L.)
- Department of Ophthalmology & Vision Science, School of Medicine, University of California Davis, Davis, CA 95616, USA
- Correspondence: ; Tel.: +1-530-752-0926
| |
Collapse
|
11
|
In silico study reveals binding potential of rotenone at multiple sites of pulmonary surfactant proteins: A matter of concern. Curr Res Toxicol 2021; 2:411-423. [PMID: 34917955 PMCID: PMC8666459 DOI: 10.1016/j.crtox.2021.11.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/20/2021] [Accepted: 11/30/2021] [Indexed: 12/21/2022] Open
Abstract
Inhalation of rotenone exposes lung surfactant proteins (SP) to this pesticide. SP-A and SP-D provides protection from microbial infection. SP-B and SP-C maintain structure and respiratory function of lungs. Rotenone has potential to bind SPs at multiple sites. Such binding can subvert functions of SPs & may invite respiratory ailments.
Rotenone is a broad-spectrum pesticide employed in various agricultural practices all over the world. Human beings are exposed to this chemical through oral, nasal, and dermal routes. Inhalation of rotenone exposes bio-molecular components of lungs to this chemical. Biophysical activity of lungs is precisely regulated by pulmonary surfactant to facilitate gaseous exchange. Surfactant proteins (SPs) are the fundamental components of pulmonary surfactant. SPs like SP-A and SP-D have antimicrobial activities providing a crucial first line of defense against infections in lungs whereas SP-B and SP-C are mainly involved in respiratory cycle and reduction of surface tension at air–water interface. In this study, molecular docking analysis using AutoDock Vina has been conducted to investigate binding potential of rotenone with the four SPs. Results indicate that, rotenone can bind with carbohydrate recognition domain (CRD) of SP-A, N-, and C- terminal peptide of SP-B, SP-C, and CRD of SP-D at multiples sites via several interaction mediators such as H bonds, C–H bonds, alkyl bonds, pi-pi stacked, Van der Waals interaction, and other. Such interactions of rotenone with SPs can disrupt biophysical and anti-microbial functions of SPs in lungs that may invite respiratory ailments and pathogenic infections.
Collapse
Key Words
- ALA, Alanine
- ARG, Arginine
- ASN, Asparagine
- ASP, Aspartic acid
- CYS, Cysteine
- Carbohydrate recognition domain
- GLN, Glutamine
- GLU, Glutamic acid
- GLY, Glycine
- HIS, Histidine
- ILE, Isoleucine
- LEU, Leucine
- LYS, Lysine
- Lungs
- MET, Methionine
- Molecular docking
- PHE, Phenylalanine
- PRO, Proline
- Rotenone
- SER, Serine
- Surfactant protein
- THR, Threonine
- TRP, Tryptophan
- TYR, Tyrosine
- VAL, Valine
Collapse
|
12
|
Sharifi S, Sharifi H, Akbari A, Dohlman CH, Paschalis EI, Gonzalez-Andrades M, Kong J, Chodosh J. Graphene-Lined Porous Gelatin Glycidyl Methacrylate Hydrogels: Implications for Tissue Engineering. ACS APPLIED NANO MATERIALS 2021; 4:12650-12662. [PMID: 35252778 PMCID: PMC8897984 DOI: 10.1021/acsanm.1c03201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Despite rigorous research, inferior mechanical properties and structural homogeneity are the main challenges constraining hydrogel's suturability to host tissue and limiting its clinical applications. To tackle those, we developed a reverse solvent interface trapping method, in which organized, graphene-coated microspherical cavities were introduced into a hydrogel to create heterogeneity and make it suturable. To generate those cavities, (i) graphite exfoliates to graphene sheets, which spread at the water/ heptane interfaces of the microemulsion, (ii) heptane fills the microspheres coated by graphene, and (iii) a cross-linkable hydrogel dissolved in water fills the voids. Cross-linking solidifies such microemulsion to a strong, suturable, permanent hybrid architecture, which has better mechanical properties, yet it is biocompatible and supports cell adhesion and proliferation. These properties along with the ease and biosafety of fabrication suggest the potential of this strategy to enhance tissue engineering outcomes by generating various suturable scaffolds for biomedical applications, such as donor cornea carriers for Boston keratoprosthesis (BK).
Collapse
Affiliation(s)
- Sina Sharifi
- Disruptive Technology Laboratory, Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Hannah Sharifi
- Disruptive Technology Laboratory, Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Ali Akbari
- Solid Tumor Research Center, Research Institute for Cellular and Molecular Medicine, Urmia University of Medical Sciences, Urmia 57147, Iran
| | - Claes H Dohlman
- Disruptive Technology Laboratory, Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Eleftherios I Paschalis
- Disruptive Technology Laboratory, Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Miguel Gonzalez-Andrades
- Disruptive Technology Laboratory, Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114, United States; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Department of Ophthalmology, Reina Sofia University Hospital and University of Cordoba, Cordoba 14004, Spain
| | - Jing Kong
- Department of Electrical Engineering andComputer Science, Massachusetts Institute of Technology,Cambridge, Massachusetts 02139, United States
| | - James Chodosh
- Disruptive Technology Laboratory, Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114, United States
| |
Collapse
|
13
|
Yang Z, Hu X, Zhou L, He Y, Zhang X, Yang J, Ju Z, Liou YC, Shen HM, Luo G, Hamblin MR, He W, Yin R. Photodynamic therapy accelerates skin wound healing through promoting re-epithelialization. BURNS & TRAUMA 2021; 9:tkab008. [PMID: 34514005 PMCID: PMC8420953 DOI: 10.1093/burnst/tkab008] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/01/2021] [Indexed: 11/16/2022]
Abstract
Background Epidermal stem cells (EpSCs) that reside in cutaneous hair follicles and the basal layer of the epidermis are indispensable for wound healing and skin homeostasis. Little is known about the effects of photochemical activation on EpSC differentiation, proliferation and migration during wound healing. The present study aimed to determine the effects of photodynamic therapy (PDT) on wound healing in vivo and in vitro. Methods We created mouse full-thickness skin resection models and applied 5-aminolevulinic acid (ALA) for PDT to the wound beds. Wound healing was analysed by gross evaluation and haematoxylin–eosin staining in vivo. In cultured EpSCs, protein expression was measured using flow cytometry and immunohistochemistry. Cell migration was examined using a scratch model; apoptosis and differentiation were measured using flow cytometry. Results PDT accelerated wound closure by enhancing EpSC differentiation, proliferation and migration, thereby promoting re-epithelialization and angiogenesis. PDT inhibited inflammatory infiltration and expression of proinflammatory cytokines, whereas the secretion of growth factors was greater than in other groups. The proportion of transient amplifying cells was significantly greater in vivo and in vitro in the PDT groups. EpSC migration was markedly enhanced after ALA-induced PDT. Conclusions Topical ALA-induced PDT stimulates wound healing by enhancing re-epithelialization, promoting angiogenesis as well as modulating skin homeostasis. This work provides a preliminary theoretical foundation for the clinical administration of topical ALA-induced PDT in skin wound healing.
Collapse
Affiliation(s)
- Zengjun Yang
- Department of Dermatology, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Xiaohong Hu
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Lina Zhou
- Department of Endocrinology, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Yaxiong He
- Department of Dermatology, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Xiaorong Zhang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Jiacai Yang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, No. 601 Huangpu Street, Tianhe District, Guangzhou, Guangdong Province, 510632, China
| | - Yih-Cherng Liou
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, 117543, Singapore
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 14 Science Drive 4, 117543, Singapore
| | - Gaoxing Luo
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, 40 Blossom Street, Boston, MA, 02114, USA
| | - Weifeng He
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Rui Yin
- Department of Dermatology, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| |
Collapse
|
14
|
Arif E, Wang C, Swiderska-Syn MK, Solanki AK, Rahman B, Manka PP, Coombes JD, Canbay A, Papa S, Nihalani D, Aspichueta P, Lipschutz JH, Syn WK. Targeting myosin 1c inhibits murine hepatic fibrogenesis. Am J Physiol Gastrointest Liver Physiol 2021; 320:G1044-G1053. [PMID: 33908271 PMCID: PMC8285590 DOI: 10.1152/ajpgi.00105.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Myosin 1c (Myo1c) is an unconventional myosin that modulates signaling pathways involved in tissue injury and repair. In this study, we observed that Myo1c expression is significantly upregulated in human chronic liver disease such as nonalcoholic steatohepatitis (NASH) and in animal models of liver fibrosis. High throughput data from the GEO-database identified similar Myo1c upregulation in mice and human liver fibrosis. Notably, transforming growth factor-β1 (TGF-β1) stimulation to hepatic stellate cells (HSCs), the liver pericyte and key cell type responsible for the deposition of extracellular matrix, upregulates Myo1c expression, whereas genetic depletion or pharmacological inhibition of Myo1c blunted TGF-β-induced fibrogenic responses, resulting in repression of α-smooth muscle actin (α-SMA) and collagen type I α 1 chain (Col1α1) mRNA. Myo1c deletion also decreased fibrogenic processes such as cell proliferation, wound healing response, and contractility when compared with vehicle-treated HSCs. Importantly, phosphorylation of mothers against decapentaplegic homolog 2 (SMAD2) and mothers against decapentaplegic homolog 3 (SMAD3) were significantly blunted upon Myo1c inhibition in GRX cells as well as Myo1c knockout (Myo1c-KO) mouse embryonic fibroblasts (MEFs) upon TGF-β stimulation. Using the genetic Myo1c-KO mice, we confirmed that Myo1c is critical for fibrogenesis, as Myo1c-KO mice were resistant to carbon tetrachloride (CCl4)-induced liver fibrosis. Histological and immunostaining analysis of liver sections showed that deposition of collagen fibers and α-SMA expression were significantly reduced in Myo1c-KO mice upon liver injury. Collectively, these results demonstrate that Myo1c mediates hepatic fibrogenesis by modulating TGF-β signaling and suggest that inhibiting this process may have clinical application in treating liver fibrosis.NEW & NOTEWORTHY The incidences of liver fibrosis are growing at a rapid pace and have become one of the leading causes of end-stage liver disease. Although TGF-β1 is known to play a prominent role in transforming cells to produce excessive extracellular matrix that lead to hepatic fibrosis, the therapies targeting TGF-β1 have achieved very limited clinical impact. This study highlights motor protein myosin-1c-mediated mechanisms that serve as novel regulators of TGF-β1 signaling and fibrosis.
Collapse
Affiliation(s)
- Ehtesham Arif
- 1Department of Medicine, Nephrology Division, Medical University of South Carolinagrid.259828.c, Charleston, South Carolina,2Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, South Carolina
| | - Cindy Wang
- 2Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, South Carolina
| | - Marzena K. Swiderska-Syn
- 3Department of Pediatrics, Darby Children’s Research Institute,
Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Ashish K. Solanki
- 1Department of Medicine, Nephrology Division, Medical University of South Carolinagrid.259828.c, Charleston, South Carolina
| | - Bushra Rahman
- 1Department of Medicine, Nephrology Division, Medical University of South Carolinagrid.259828.c, Charleston, South Carolina
| | - Paul P. Manka
- 2Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, South Carolina,4Department of Medicine, University Hospital Knappschaftskrankenhaus, Ruhr-University Bochum, Bochum, Germany
| | - Jason D. Coombes
- 5Institute of Hepatology, Foundation for Liver Research, London, United Kingdom,6School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Ali Canbay
- 4Department of Medicine, University Hospital Knappschaftskrankenhaus, Ruhr-University Bochum, Bochum, Germany
| | - Salvatore Papa
- 7Leeds Institute of Medical Research at St. James’s, Faculty of
Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Deepak Nihalani
- 1Department of Medicine, Nephrology Division, Medical University of South Carolinagrid.259828.c, Charleston, South Carolina,8Division of Kidney, Urologic and Hematologic Diseases, National Institutes of Health, Bethesda, Maryland
| | - Patricia Aspichueta
- 9Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), Leioa, Spain
| | - Joshua H. Lipschutz
- 1Department of Medicine, Nephrology Division, Medical University of South Carolinagrid.259828.c, Charleston, South Carolina,10Section of Nephrology, Ralph H Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Wing-Kin Syn
- 2Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, South Carolina,9Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), Leioa, Spain,11Section of Gastroenterology, Ralph H Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| |
Collapse
|
15
|
García-Posadas L, Diebold Y. Three-Dimensional Human Cell Culture Models to Study the Pathophysiology of the Anterior Eye. Pharmaceutics 2020; 12:E1215. [PMID: 33333869 PMCID: PMC7765302 DOI: 10.3390/pharmaceutics12121215] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/11/2020] [Accepted: 12/13/2020] [Indexed: 02/07/2023] Open
Abstract
In recent decades, the establishment of complex three-dimensional (3D) models of tissues has allowed researchers to perform high-quality studies and to not only advance knowledge of the physiology of these tissues but also mimic pathological conditions to test novel therapeutic strategies. The main advantage of 3D models is that they recapitulate the spatial architecture of tissues and thereby provide more physiologically relevant information. The eye is an extremely complex organ that comprises a large variety of highly heterogeneous tissues that are divided into two asymmetrical portions: the anterior and posterior segments. The anterior segment consists of the cornea, conjunctiva, iris, ciliary body, sclera, aqueous humor, and the lens. Different diseases in these tissues can have devastating effects. To study these pathologies and develop new treatments, the use of cell culture models is instrumental, and the better the model, the more relevant the results. Thus, the development of sophisticated 3D models of ocular tissues is a significant challenge with enormous potential. In this review, we present a comprehensive overview of the latest advances in the development of 3D in vitro models of the anterior segment of the eye, with a special focus on those that use human primary cells.
Collapse
Affiliation(s)
- Laura García-Posadas
- Instituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, 47011 Valladolid, Spain;
| | - Yolanda Diebold
- Instituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, 47011 Valladolid, Spain;
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
16
|
Gonzalez-Andrades M, Jalimarada SS, Rodriguez-Benavente M, Feeley MN, Woodward AM, AbuSamra DB, Argüeso P. Golgi α1,2-mannosidase I induces clustering and compartmentalization of CD147 during epithelial cell migration. Cell Adh Migr 2020; 14:96-105. [PMID: 32419574 PMCID: PMC7250185 DOI: 10.1080/19336918.2020.1764170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 03/08/2020] [Accepted: 04/27/2020] [Indexed: 01/05/2023] Open
Abstract
CD147 is a widely expressed matrix metalloproteinase inducer involved in the regulation of cell migration. The high glycosylation and ability to undergo oligomerization have been linked to CD147 function, yet there is limited understanding on the molecular mechanisms behind these processes. The current study demonstrates that the expression of Golgi α1,2-mannosidase I is key to maintaining the cell surface organization of CD147 during cell migration. Using an in vitro model of stratified human corneal epithelial wound healing, we show that CD147 is clustered within lateral plasma membranes at the leading edge of adjacent migrating cells. This localization correlates with a surge in matrix metalloproteinase activity and an increase in the expression of α1,2-mannosidase subtype IC (MAN1C1). Global inhibition of α1,2-mannosidase I activity with deoxymannojirimycin markedly attenuates the glycosylation of CD147 and disrupts its surface distribution at the leading edge, concomitantly reducing the expression of matrix metalloproteinase-9. Likewise, treatment with deoxymannojirimycin or siRNA-mediated knockdown of MAN1C1 impairs the ability of the carbohydrate-binding protein galectin-3 to stimulate CD147 clustering in unwounded cells. We conclude that the mannose-trimming activity of α1,2-mannosidase I coordinates the clustering and compartmentalization of CD147 that follows an epithelial injury.
Collapse
Affiliation(s)
- Miguel Gonzalez-Andrades
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Supriya S. Jalimarada
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Maria Rodriguez-Benavente
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Marissa N. Feeley
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Ashley M. Woodward
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Dina B. AbuSamra
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Pablo Argüeso
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Gahane AY, Singh V, Kumar A, Kumar Thakur A. Development of mechanism-based antibacterial synergy between Fmoc-phenylalanine hydrogel and aztreonam. Biomater Sci 2020; 8:1996-2006. [PMID: 32073033 DOI: 10.1039/c9bm01978b] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recently, fluorenylmethyloxycarbonyl (Fmoc) conjugated amino acids (Fmoc-AA), especially Fmoc-phenylalanine (Fmoc-F), have been discovered to have antimicrobial properties specific to Gram-positive bacteria including MRSA. Their weak antibacterial activity against Gram-negative bacteria is due to their inability to cross the bacterial membrane. Here in order to increase the antibacterial spectrum of Fmoc-F, we prepared a formulation of Fmoc-F with the Gram-negative specific antibiotic aztreonam (AZT). This formulation displayed antibacterial activity against both Gram-positive and Gram-negative bacteria and significantly reduced the bacterial load in a mouse wound infection model. The combination produced a synergistic effect and higher efficacy against P. aeruginosa due to the increased Fmoc-F permeability by AZT through the bacterial membrane. This combinatorial approach could be an effective strategy for other Fmoc-AA having a Gram-positive specific antibacterial effect for the better management of bacterial wound infections.
Collapse
Affiliation(s)
- Avinash Yashwant Gahane
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India.
| | - Virender Singh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India.
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India.
| | - Ashwani Kumar Thakur
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India.
| |
Collapse
|
18
|
Yang GN, Strudwick XL, Bonder C, Kopecki Z, Cowin AJ. Effect of Flightless I Expression on Epidermal Stem Cell Niche During Wound Repair. Adv Wound Care (New Rochelle) 2020; 9:161-173. [PMID: 32117580 DOI: 10.1089/wound.2018.0884] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 06/03/2019] [Indexed: 12/31/2022] Open
Abstract
Objective: Activation of epidermal stem cells (EpSCs) from their quiescent niche is an integral component of wound reepithelialization and involves Wnt/β-catenin (β-Cat) signaling and remodeling of the actin cytoskeleton. The aim of this study was to investigate the effect of Flightless I (Flii), a cytoskeletal protein and inhibitor of wound healing, on EpSC activation during wound repair. Approach: Genetically modified Flii mice (Flii knockdown: Flii+/- , wild type: WT, Flii overexpressing: FliiTg/Tg ) received two incisional wounds along the lateral axis of the dorsal skin. Indicators of EpSC activation (epidermal growth factor receptor 1 [EGFR1], leucine-rich repeats and immunoglobulin-like domains-1 [Lrig1], K14), Wnt/β-Cat signaling (Lgr6, Flap2, β-Cat, and axis inhibition protein 2 [Axin2]), and cell proliferation (proliferating cell nuclear antigen [PCNA]) were assessed using immunohistochemistry. β-Cat stabilization was examined using western blotting with cell cycling and differentiation of isolated CD34+ITGA6high EpSCs examined using real time-quantitative polymerase chain reaction after treatment with wound-conditioned media. Results: Flii+/- led to increased numbers of activated EpSCs expressing PCNA, elevated EGFR1, and decreased Lrig1. EpSCs in Flii+/- hair follicle niches adjacent to the wounds also showed expression of Wnt-activation markers including increased β-Cat and Lgr6, and decreased Axin2. EpSCs (CD34+ITGA6high) isolated from Flii+/- unwounded skin showed elevated expression of cell-cycling genes including ΔNp63, filaggrin (Fila), involucrin (Invo), cyclin D1 (Ccnd1), and cell-division cycle protein-20 (Cdc20); and elevated ΔNp63 and Invo after treatment with wound-conditioned media compared with WT and FliiTg/Tg counterparts. Innovation: Flii was identified as an inhibitor of EpSC activation that may explain its negative effects on wound reepithelialization. Conclusion: Flii may inhibit EpSC activation by interrupting Wnt/β-Cat signaling. Strategies that reduce Flii may increase activation of EpSCs and promote reepithelialization of wounds.
Collapse
Affiliation(s)
- Gink N. Yang
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - Xanthe L. Strudwick
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - Claudine Bonder
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Zlatko Kopecki
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - Allison J. Cowin
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
19
|
Kaluzhny Y, Kinuthia MW, Lapointe AM, Truong T, Klausner M, Hayden P. Oxidative stress in corneal injuries of different origin: Utilization of 3D human corneal epithelial tissue model. Exp Eye Res 2019; 190:107867. [PMID: 31705899 DOI: 10.1016/j.exer.2019.107867] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/10/2019] [Accepted: 11/04/2019] [Indexed: 12/17/2022]
Abstract
The purpose of the current work was to utilize a three dimensional (3D) corneal epithelial tissue model to study dry eye disease and oxidative stress-related corneal epithelial injuries for the advancement of ocular therapeutics. Air-liquid interface cultures of normal human corneal epithelial cells were used to produce 3D corneal epithelial tissues appropriate for physiologically relevant exposure to environmental factors. Oxidative stress was generated by exposing the tissues to non-toxic doses of ultraviolet radiation (UV), hydrogen peroxide, vesicating agent nitrogen mustard, or desiccating conditions that stimulated morphological, cellular, and molecular changes relevant to dry eye disease. Corneal specific responses, including barrier function, tissue viability, reactive oxygen species (ROS) accumulation, lipid peroxidation, cytokine release, histology, and gene expression were evaluated. 3D corneal epithelial tissue model structurally and functionally reproduced key features of molecular responses of various types of oxidative stress-induced ocular damage. The most pronounced effects for different treatments were: UV irradiation - intracellular ROS accumulation; hydrogen peroxide exposure - barrier impairment and IL-8 release; nitrogen mustard exposure - lipid peroxidation and IL-8 release; desiccating conditions - tissue thinning, a decline in mucin expression, increased lipid peroxidation and IL-8 release. Utilizing a PCR gene array, we compared the effects of corneal epithelial damage on the expression of 84 oxidative stress-responsive genes and found specific molecular responses for each type of damage. The topical application of lubricant eye drops improved tissue morphology while decreasing lipid peroxidation and IL-8 release from tissues incubated at desiccating conditions. This model is anticipated to be a valuable tool to study molecular mechanisms of corneal epithelial damage and aid in the development of therapies against dry eye disease, oxidative stress- and vesicant-induced ocular injuries.
Collapse
Affiliation(s)
- Yulia Kaluzhny
- MatTek Corporation, 200 Homer Avenue, Ashland, MA, 01721, USA.
| | | | | | - Thoa Truong
- MatTek Corporation, 200 Homer Avenue, Ashland, MA, 01721, USA.
| | | | - Patrick Hayden
- MatTek Corporation, 200 Homer Avenue, Ashland, MA, 01721, USA.
| |
Collapse
|
20
|
Kiesewetter L, Littau L, Walles H, Boccaccini AR, Groeber-Becker F. Reepithelialization in focus: Non-invasive monitoring of epidermal wound healing in vitro. Biosens Bioelectron 2019; 142:111555. [DOI: 10.1016/j.bios.2019.111555] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 01/10/2023]
|
21
|
Islam MM, Sharifi R, Mamodaly S, Islam R, Nahra D, Abusamra DB, Hui PC, Adibnia Y, Goulamaly M, Paschalis EI, Cruzat A, Kong J, Nilsson PH, Argüeso P, Mollnes TE, Chodosh J, Dohlman CH, Gonzalez-Andrades M. Effects of gamma radiation sterilization on the structural and biological properties of decellularized corneal xenografts. Acta Biomater 2019; 96:330-344. [PMID: 31284096 PMCID: PMC7043233 DOI: 10.1016/j.actbio.2019.07.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/21/2019] [Accepted: 07/03/2019] [Indexed: 12/19/2022]
Abstract
To address the shortcomings associated with corneal transplants, substantial efforts have been focused on developing new modalities such as xenotransplantion. Xenogeneic corneas are anatomically and biomechanically similar to the human cornea, yet their applications require prior decellularization to remove the antigenic components to avoid rejection. In the context of bringing decellularized corneas into clinical use, sterilization is a crucial step that determines the success of the transplantation. Well-standardized sterilization methods, such as gamma irradiation (GI), have been applied to decellularized porcine corneas (DPC) to avoid graft-associated infections in human recipients. However, little is known about the effect of GI on decellularized corneal xenografts. Here, we evaluated the radiation effect on the ultrastructure, optical, mechanical and biological properties of DPC. Transmission electron microscopy revealed that gamma irradiated decellularized porcine cornea (G-DPC) preserved its structural integrity. Moreover, the radiation did not reduce the optical properties of the tissue. Neither DPC nor G-DPC led to further activation of complement system compared to native porcine cornea when exposed to plasma. Although, DPC were mechanically comparable to the native tissue, GI increased the mechanical strength, tissue hydrophobicity and resistance to enzymatic degradation. Despite these changes, human corneal epithelial, stromal, endothelial and hybrid neuroblastoma cells grew and differentiated on DPC and G-DPC. Thus, GI may achieve effective tissue sterilization without affecting critical properties that are essential for corneal transplant survival.
Collapse
Affiliation(s)
- Mohammad Mirazul Islam
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Roholah Sharifi
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Shamina Mamodaly
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Rakibul Islam
- Department of Immunology, Oslo University Hospital, Rikshospitalet, University of Oslo, Oslo, Norway
| | - Daniel Nahra
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Dina B Abusamra
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Pui Chuen Hui
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Yashar Adibnia
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Yeditepe University School of Medicine, Istanbul, Turkey
| | - Mehdi Goulamaly
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Eleftherios I Paschalis
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Andrea Cruzat
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Department of Ophthalmology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jing Kong
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Per H Nilsson
- Department of Immunology, Oslo University Hospital, Rikshospitalet, University of Oslo, Oslo, Norway; Linnaeus Center for Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Pablo Argüeso
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Tom Eirik Mollnes
- Department of Immunology, Oslo University Hospital, Rikshospitalet, University of Oslo, Oslo, Norway; Research Laboratory, Nordland Hospital, Bodø, and Faculty of Health Sciences, K.G. Jebsen TREC, University of Tromsø, Norway; Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - James Chodosh
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Claes H Dohlman
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Miguel Gonzalez-Andrades
- Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Department of Ophthalmology, Reina Sofia University Hospital and University of Cordoba, Cordoba, Spain.
| |
Collapse
|
22
|
Slater B, Ng E, McGuigan AP. Local cell coordination does not alter individual cell migration during collective migration but does impact cellular exchange events. Integr Biol (Camb) 2019; 11:163-172. [DOI: 10.1093/intbio/zyz015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/11/2019] [Accepted: 05/12/2019] [Indexed: 01/23/2023]
Abstract
Abstract
Coordinated cell re-organization is critical to ensure correct tissue morphogenesis for a number of important embryonic and tissue repair events, however the mechanisms that govern cells coordination during collective movements, particularly in situations where cells are spatially restricted by their neighbours, are not well understood. Here we assessed cell re-organization in monolayers of retinal epithelial cells (ARPE-19) to determine if cells that coordinate with their neighbours exhibit differential migration properties to non-coordinating cells and participate differently in local cell re-organization of the tissue sheet. From global tracking analysis, we determined that the movement profiles of cells were indistinguishable regardless of whether or not they were a part of multicellular streams. Using high magnification live imaging of cell membranes, we also characterized the localized geometry and organization of a monolayer (cell area, number of nearest neighbours, aspect ratio, internal cell angles) during cell re-organization in both streaming and non-streaming regions. Consistent with our global migration analysis, we observed no differences in cell sheet geometry and organization in streaming versus non-streaming regions. We did however observe that cells executed T1-like transitions to exchange position within the space-limited monolayer and that exchange events consistently involved at least one non-streaming cell. Our data suggests a model in which cell movement within the sheet is limited by neighbour exchange events and likely cells transition between streaming and non-streaming regimes to facilitate these neighbour exchange events while maintaining the integrity of the sheet.
Collapse
Affiliation(s)
- Benjamin Slater
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St., Toronto, ON, Canada
| | - Edwin Ng
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St., Toronto, ON, Canada
| | - Alison P McGuigan
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St., Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, Canada
| |
Collapse
|
23
|
Scholler J, Mazlin V, Thouvenin O, Groux K, Xiao P, Sahel JA, Fink M, Boccara C, Grieve K. Probing dynamic processes in the eye at multiple spatial and temporal scales with multimodal full field OCT. BIOMEDICAL OPTICS EXPRESS 2019; 10:731-746. [PMID: 30800511 PMCID: PMC6377896 DOI: 10.1364/boe.10.000731] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/07/2019] [Accepted: 01/07/2019] [Indexed: 05/08/2023]
Abstract
We describe recent technological progress in multimodal en face full-field optical coherence tomography that has allowed detection of slow and fast dynamic processes in the eye. We show that by combining static, dynamic and fluorescence contrasts we can achieve label-free high-resolution imaging of the retina and anterior eye with temporal resolution from milliseconds to several hours, allowing us to probe biological activity at subcellular scales inside 3D bulk tissue. Our setups combine high lateral resolution over a large field of view with acquisition at several hundreds of frames per second which make it a promising tool for clinical applications and biomedical studies. Its contactless and non-destructive nature is shown to be effective for both following in vitro sample evolution over long periods of time and for imaging of the human eye in vivo.
Collapse
Affiliation(s)
- Jules Scholler
- Institut Langevin, ESPCI Paris, CNRS, PSL University, 1 rue Jussieu, 75005 Paris,
France
| | - Viacheslav Mazlin
- Institut Langevin, ESPCI Paris, CNRS, PSL University, 1 rue Jussieu, 75005 Paris,
France
| | - Olivier Thouvenin
- Institut Langevin, ESPCI Paris, CNRS, PSL University, 1 rue Jussieu, 75005 Paris,
France
| | - Kassandra Groux
- Institut Langevin, ESPCI Paris, CNRS, PSL University, 1 rue Jussieu, 75005 Paris,
France
- LLTech SAS, 29 Rue du Faubourg Saint Jacques, Paris, 75014,
France
| | - Peng Xiao
- Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou,
China
| | - José-Alain Sahel
- Vision Institute/CIC 1423, UPMC-Sorbonne Universities, UMR_S 968/INSERM, U968/CNRS, UMR_7210, 17 Rue Moreau, Paris, 75012,
France
- Quinze-Vingts National Eye Hospital, 28 Rue de Charenton, Paris, 75012,
France
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213,
USA
| | - Mathias Fink
- Institut Langevin, ESPCI Paris, CNRS, PSL University, 1 rue Jussieu, 75005 Paris,
France
| | - Claude Boccara
- Institut Langevin, ESPCI Paris, CNRS, PSL University, 1 rue Jussieu, 75005 Paris,
France
- LLTech SAS, 29 Rue du Faubourg Saint Jacques, Paris, 75014,
France
| | - Kate Grieve
- Vision Institute/CIC 1423, UPMC-Sorbonne Universities, UMR_S 968/INSERM, U968/CNRS, UMR_7210, 17 Rue Moreau, Paris, 75012,
France
- Quinze-Vingts National Eye Hospital, 28 Rue de Charenton, Paris, 75012,
France
| |
Collapse
|
24
|
Improving the practicality and safety of artificial corneas: Pre-assembly and gamma-rays sterilization of the Boston Keratoprosthesis. Ocul Surf 2018; 16:322-330. [PMID: 29627599 DOI: 10.1016/j.jtos.2018.04.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/03/2018] [Accepted: 04/04/2018] [Indexed: 11/22/2022]
Abstract
PURPOSE To make the Boston keratoprosthesis (B-KPro), together with its carrier corneal graft, more easily procured, transported and stored, as well as less expensive, easier for the surgeon to implant and safer for the patient, it is proposed that the B-KPro-graft combination be pre-assembled by an expert technician, followed by sterilization with gamma ray irradiation (GI) allowing long-term storage at room temperature. For this to be possible, it must be shown that the B-KPro itself (not only the graft) remains unharmed by the irradiation. METHODS Polymethyl methacrylate (PMMA) discs and B-KPros were submitted to either ethylene oxide sterilization or different doses of GI. Cell biocompatibility, mechanical strength and optical quality were evaluated. The feasibility of assembling the B-KPro to a corneal graft, and gamma-radiate afterwards, was also assessed. RESULTS There were no differences in cell biocompatibility between the samples. The optical evaluation showed high levels of transparency for all the groups. The absorbance of ultraviolet was higher for the groups treated with GI. The mechanical evaluation by nanoindentation showed no alterations of the PMMA discs after GI. The flexure test revealed a similar mechanical behavior. Technically, pre-assembly and GI of the B-KPro revealed no problems. CONCLUSIONS Sterilization of B-KPro using GI has no detrimental influence on the device. The pre-assembly of B-KPro to a donor cornea, followed by gamma sterilization, emerges as an efficient and safe procedure.
Collapse
|
25
|
Sharif R, Priyadarsini S, Rowsey TG, Ma JX, Karamichos D. Corneal Tissue Engineering: An In Vitro Model of the Stromal-nerve Interactions of the Human Cornea. J Vis Exp 2018. [PMID: 29443018 DOI: 10.3791/56308] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Tissue engineering has gained substantial recognition due to the high demand for human cornea replacements with an estimated 10 million people worldwide suffering from corneal vision loss1. To address the demand for viable human corneas, significant progress in three-dimensional (3D) tissue engineering has been made2,3,4. These cornea models range from simple monolayer systems to multilayered models, leading to 3D full-thickness corneal equivalents2. However, the use of a 3D tissue-engineered cornea in the context of in vitro disease models studied to date lacks resemblance to the multilayered 3D corneal tissue structure, function, and the networking of different cell types (i.e., nerve, epithelium, stroma, and endothelium)2,3. In addition, the demand for in vitro cornea tissue models has increased in an attempt to reduce animal testing for pharmaceutical products. Thus, more sophisticated models are required to better match systems to human physiological requirements, and the development of a model that is more relevant to the patient population is absolutely necessary. Given that multiple cell types in the cornea are affected by diseases and dystrophies, such as Keratoconus, Diabetic Keratopathy, and Fuchs, this model includes a 3D co-culture model of primary human corneal fibroblasts (HCFs) from healthy donors and neurons from the SH-SY5Y cell line. This allows us for the first time to investigate the interactions between the two cell types within the human corneal tissue. We believe that this model could potentially dissect the underlying mechanisms associated with the stromal-nerve interactions of corneal diseases that exhibit nerve damages. This 3D model mirrors the basic anatomical and physiological nature of the corneal tissue in vivo and can be used in the future as a tool for investigating corneal defects as well as screening the efficacy of various agents before animal testing.
Collapse
Affiliation(s)
- Rabab Sharif
- Department of Cell Biology, University of Oklahoma Health Sciences Center
| | - Shrestha Priyadarsini
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center
| | - Tyler G Rowsey
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center
| | - Jian-Xing Ma
- Department of Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center
| | - Dimitrios Karamichos
- Department of Cell Biology, University of Oklahoma Health Sciences Center; Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center;
| |
Collapse
|
26
|
Priyadarsini S, Nicholas SE, Karamichos D. 3D Stacked Construct: A Novel Substitute for Corneal Tissue Engineering. Methods Mol Biol 2017; 1697:173-180. [PMID: 28451994 DOI: 10.1007/7651_2017_23] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Corneal trauma/injury often results in serious complications including permanent vision loss or loss of visual acuity which demands corneal transplantations or treatment with allogenic graft tissues. There is currently a huge shortage of donor tissue worldwide and the need for human corneal equivalents increases annually. In order to meet such demand the current clinical approach of treating corneal injuries is limited and involves synthetic and allogenic materials which have various shortcomings when it comes to actual transplantations. In this study we introduce the newly developed, next generation of our previously established 3D self-assembled constructs, where multiple constructs are grown and stacked on top of each other without any other artificial product. This new technology brings our 3D in vitro model closer to what is seen in vivo and provides a solid foundation for future studies on corneal biology.Lipids are known for playing a vital role during metabolism and diseased state of various tissues and Sphingolipids are one such class of lipids which are involved in various cellular mechanisms and signaling processes. The impacts of Sphingolipids that have been documented in several human diseases often involve inflammation, neovascularization, tumorigenesis, and diabetes, but these conditions are not yet thoroughly studied. There is very little information about the exact role of Sphingolipids in the human cornea and future studies aiming at dissecting the mechanisms and pathways involved in order to develop novel therapies. We believe that our novel 3D stacked model can be used to delineate the role of Sphingolipids in the human cornea and provide new insights for understanding and treating various human corneal diseases.
Collapse
Affiliation(s)
- Shrestha Priyadarsini
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Sarah E Nicholas
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Dimitrios Karamichos
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
27
|
Begnaud S, Chen T, Delacour D, Mège RM, Ladoux B. Mechanics of epithelial tissues during gap closure. Curr Opin Cell Biol 2016; 42:52-62. [PMID: 27131272 DOI: 10.1016/j.ceb.2016.04.006] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 04/06/2016] [Accepted: 04/08/2016] [Indexed: 12/15/2022]
Abstract
The closure of gaps is crucial to maintaining epithelium integrity during developmental and repair processes such as dorsal closure and wound healing. Depending on biochemical as well as physical properties of the microenvironment, gap closure occurs through assembly of multicellular actin-based contractile cables and/or protrusive activity of cells lining the gap. This review discusses the relative contributions of 'purse-string' and cell crawling mechanisms regulated by cell-substrate and cell-cell interactions, cellular mechanics and physical constraints from the environment.
Collapse
Affiliation(s)
- Simon Begnaud
- Institut Jacques Monod (IJM), CNRS UMR 7592 & University Paris Diderot, Paris, France
| | - Tianchi Chen
- Mechanobiology Institute (MBI), National University of Singapore, Singapore
| | - Delphine Delacour
- Institut Jacques Monod (IJM), CNRS UMR 7592 & University Paris Diderot, Paris, France
| | - René-Marc Mège
- Institut Jacques Monod (IJM), CNRS UMR 7592 & University Paris Diderot, Paris, France.
| | - Benoît Ladoux
- Institut Jacques Monod (IJM), CNRS UMR 7592 & University Paris Diderot, Paris, France; Mechanobiology Institute (MBI), National University of Singapore, Singapore.
| |
Collapse
|