Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.015
Filter
Add more filters

Publication year range
1.
Curr Opin Ophthalmol ; 35(4): 309-314, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38813737

ABSTRACT

PURPOSE OF REVIEW: To highlight the progress and future direction of limbal stem cell (LSC) therapies for the treatment of limbal stem cell deficiency (LSCD). RECENT FINDINGS: Direct LSC transplantation have demonstrated good long-term outcomes. Cultivated limbal epithelial transplantation (CLET) has been an alternative to treat severe to total LSCD aiming to improve the safety and efficacy of the LSC transplant. A prospective early-stage uncontrolled clinical trial shows the feasibility and safety of CLET manufactured under xenobiotic free conditions. Other cell sources for repopulating of the corneal epithelium such as mesenchymal stem cells (MSCs) and induced pluripotent stem cells are being investigated. The first clinical trials of using MSCs showed short-term results, but long-term efficacy seems to be disappointing. A better understanding of the niche function and regulation of LSC survival and proliferation will lead to the development of medical therapies to rejuvenate the residual LSCs found in a majority of eyes with LSCD in vivo. Prior efforts have been largely focused on improving LSC transplantation. Additional effort should be placed on improving the accuracy of diagnosis and staging of LSCD, and implementing standardized outcome measures which enable comparison of efficacy of different LSCD treatments for different severity of LSCD. The choice of LSCD treatment will be customized based on the severity of LSCD in the future. SUMMARY: New approaches for managing different stages of LSCD are being developed. This concise review summarizes the progresses in LSC therapies for LSCD, underlying mechanisms, limitations, and future areas of development.


Subject(s)
Corneal Diseases , Limbus Corneae , Stem Cell Transplantation , Humans , Limbus Corneae/cytology , Stem Cell Transplantation/methods , Corneal Diseases/therapy , Corneal Diseases/surgery , Epithelium, Corneal , Limbal Stem Cells
2.
Int Ophthalmol ; 44(1): 337, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39093517

ABSTRACT

PURPOSE: To evaluate limbal graft transplantation success in pediatric patients with chemical injury-induced limbal stem cell deficiency (LSCD) using the 'LSCD Working Group' staging system. METHODS: Medical records of 11 eyes of 11 children who underwent limbal graft transplantation (limbal autograft/limbal allograft) were included. Surgical success was defined as improvement in the post-operative 1st year LSCD stage. RESULTS: The mean age was 12 ± 5 (4-17) years. Causative agent was alkaline in 4(36.4%) and acid in 3(27.2%) patients. Limbal autograft was performed in 9 (81.8%) eyes with unilateral LSCD, and allograft transplantation was performed in 2 (18.2%) eyes with bilateral LSCD. The mean follow-up time was 33.89 ± 30.73 (12-102.33) months. The overall limbal graft transplantation success rate was 72.7%. Among 9 patients who receive limbal autograft, 8 had improvement in post-operative LSCD stage, 1 had stable LSCD stage. Of the 2 patients who receive limbal allograft, post-operative LSCD stage remained the same in 1 and worsened in 1 patient. The mean time between injury and the surgery was 30.47 ± 30.08 (7-108.47) months. Penetrating keratoplasty was performed in 3 (27.2%) of 11 patients following limbal graft transplantation. CONCLUSION: Management of LSCD in children is challenging and appears to be somewhat different from that of adults. Limited data in the literature indicate that cultivated or simple limbal epithelial transplantations (CLET/SLET) are primarily preferred in children. Although the tendency to take small tissue from the healthy eye is noteworthy, conventional limbal allograft and autograft transplantations also show promising results without any further complications in at least 1 year follow-up period.


Subject(s)
Limbal Stem Cell Deficiency , Limbus Corneae , Visual Acuity , Adolescent , Child , Child, Preschool , Female , Humans , Male , Burns, Chemical/surgery , Corneal Transplantation/methods , Eye Burns/surgery , Eye Burns/chemically induced , Eye Burns/diagnosis , Follow-Up Studies , Limbal Stem Cell Deficiency/chemically induced , Limbal Stem Cell Deficiency/diagnosis , Limbal Stem Cell Deficiency/surgery , Limbus Corneae/cytology , Retrospective Studies , Stem Cell Transplantation/methods , Stem Cells/cytology , Transplantation, Autologous , Treatment Outcome
3.
Vestn Oftalmol ; 140(2. Vyp. 2): 80-89, 2024.
Article in Russian | MEDLINE | ID: mdl-38739135

ABSTRACT

Limbal stem cell deficiency (LSCD) is one of the leading factors negatively affecting the success of keratoplasty, and its treatment remains an urgent problem in ophthalmology. With the development of regenerative medicine, one of the promising approaches is the transplantation of tissue-engineered constructs from cultured limbal stem cells (LSCs) in biopolymer carriers. PURPOSE: This study was conducted to develop an experimental model of LSCD and evaluate the effectiveness of transplantation of a tissue-engineered construct consisting of cultured cells containing a population of LSCs and a collagen carrier. MATERIAL AND METHODS: The study was performed on 12 rabbits and included several stages. At the first stage, the physiological effects of collagen matrix implantation into the limbal zone were studied. At the second stage, tissue-engineered constructs consisting of LSCs on a collagen matrix were formed and their effect on the regeneration processes in the experimental LSCD model was analyzed. The animals were divided into 2 groups: surgical treatment (transplantation of the tissue-engineered construct) was used in the experimental group, and conservative treatment was used in the control group. Slit-lamp biomicroscopy with photo-registration, fluorescein corneal staining, optical coherence tomography of the anterior segment of the eye, and impression cytology were used to assess the results. RESULTS: No side reactions were observed after implantation of the collagen matrix into the limbal zone. One month after surgical treatment of the LSCD model in the experimental group, complete epithelization with minor manifestations of epitheliopathy was observed. In the control group, erosion of the corneal epithelium was noted. The time of corneal epithelization in the experimental and control groups was 9.2±2.95 and 46.20±12.07 days, respectively (p=0.139). According to the data of impression cytology, in the experimental group there were no goblet cells in the central part of the cornea, which indicates the restoration of corneal type epithelial cells, in contrast to the control group. CONCLUSION: Transplantation of a tissue-engineered construct from cultured limbal cells on a collagen membrane should be considered as a promising method for the treatment of limbal stem cell deficiency.


Subject(s)
Corneal Diseases , Disease Models, Animal , Limbus Corneae , Stem Cell Transplantation , Stem Cells , Tissue Engineering , Rabbits , Animals , Tissue Engineering/methods , Limbus Corneae/cytology , Corneal Diseases/therapy , Corneal Diseases/surgery , Stem Cell Transplantation/methods , Cells, Cultured , Tomography, Optical Coherence/methods , Treatment Outcome , Limbal Stem Cell Deficiency
4.
Stem Cells ; 39(3): 280-295, 2021 03.
Article in English | MEDLINE | ID: mdl-33373496

ABSTRACT

Quiescence and self-renewal of human corneal epithelial progenitor/stem cells (LEPC) are regulated by the limbal niche, presumably through close interaction with limbal (stromal) niche cells (LNC). Paired box homeotic gene 6 (Pax6), a conserved transcription factor essential for eye development, is essential for proper differentiation of limbal and corneal epithelial stem cells. Pax6 haploinsufficiency causes limbal stem cell deficiency, which leads to subsequent corneal blindness. We previously reported that serial passage of nuclear Pax6+ LNC resulted in the gradual loss of nuclear Pax6+ and neural crest progenitor status, the latter of which was reverted upon recovery of Pax6. These findings suggest Pax6 plays a pivotal role in supporting the self-renewal of LEPC in limbal niche. Herein, we show that HC-HA/PTX3, a unique matrix purified from amniotic membrane (AM) and consists of heavy chain 1of inter-α-trypsin inhibitor covalently linked to hyaluronic acid and complexed with pentraxin 3, is capable of reverting senescent LNC to nuclear Pax6+ neural crest progenitors that support self-renewal of LEPC. Such reversion is causally linked to early cell aggregation mediated by activation of C-X-C chemokine receptor type 4 (CXCR4)-mediated signaling followed by activation of bone morphogenetic protein (BMP) signaling. Furthermore, CXCR4-mediated signaling, but not BMP signaling, controls recovery of the nuclear Pax6+ neural crest progenitors. These findings not only explain why AM helps in vivo and ex vivo expansion of human LEPC, but they also illuminate the potential role of HC-HA/PTX3 as a surrogate matrix niche that complements stem cell-based therapies in regenerative medicine.


Subject(s)
C-Reactive Protein/metabolism , Limbus Corneae/cytology , PAX6 Transcription Factor/metabolism , Serum Amyloid P-Component/metabolism , Stem Cell Niche/physiology , Aged , Cell Differentiation/physiology , Cells, Cultured , Corneal Diseases/genetics , Epithelial Cells/metabolism , Epithelium, Corneal/cytology , Humans , Middle Aged , Neural Crest/cytology , Stem Cells/metabolism
5.
Mol Vis ; 27: 370-383, 2021.
Article in English | MEDLINE | ID: mdl-34447239

ABSTRACT

Purpose: Viral infections such as herpetic keratitis (HSK) activate the innate immune response in the cornea triggering opacity and loss of vision. This condition is performed mainly by myofibroblasts that exacerbate secretion of inflammatory cytokines. Amniotic membrane transplantation (AMT) reduces ocular opacity and scarring inhibiting secretion of inflammatory cytokines and proliferation of myofibroblasts. We previously reported that the amniotic membrane (AM) favors an anti-inflammatory microenvironment inhibiting the secretion of inflammatory cytokines, expression of innate immune receptors, and translocation of nuclear NF-κB on human limbal myofibroblasts (HLMs). The aim of the present study was to determine whether the soluble factors of the AM decrease the immune response of HLMs stimulated with polyinosinic-polycytidylic acid sodium salt (poly I:C). Methods: The AM was incubated in Dulbecco's modified eagle medium (DMEM)/F12, and the supernatant was collected to obtain amniotic membrane conditioned medium (AMCM). HLMs were isolated from cadaveric sclera-corneal rims. HLMs were cultured in DMEM/F12 or AMCM and stimulated or not with poly I:C (10 µg/ml) for 12 h to analyze synthesis of CCL2, CCL5, CXCL10, MDA5, RIG-1, and TLR3 or for 2 h to analyze translocation of nuclear NF-kB, IRF3, and IRF7. The proteins contained on AMCM were analyzed by matrix assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS), and the acquired peptide ions were analyzed with the Mascot program using both National Center for Biotechnology Information (NCBI) and expressed sequence tag (EST) databases. Results: AMCM downregulated the mRNA levels of CCL2, CCL5, CXCL10, MDA5, RIG-1, and TLR3. In addition, AMCM decreased secretion of CCL2, CCL5, and CXCL10 and translocation of nuclear NF-κB. Interestingly, AMCM increased translocation of nuclear IRF3 and synthesis and secretion of type I IFN-ß. We also identified small leucine-rich proteoglycan lumican in the AMCM. The administration of rh-lumican to poly I:C-stimulated HLMs reduced the mRNA levels of CCL2, CCL5, and CXCL10. Conclusions: These results suggest that the AM can trigger an anti-inflammatory response on HLMs through soluble factors, and that lumican could play an important role in these effects.


Subject(s)
Amnion/physiology , Culture Media, Conditioned/pharmacology , Inflammation/prevention & control , Limbus Corneae/cytology , Myofibroblasts/drug effects , Cells, Cultured , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique, Indirect , Humans , Immunity, Innate/drug effects , Lumican/pharmacology , Myofibroblasts/metabolism , NF-kappa B/metabolism , Phosphorylation , Poly I-C/pharmacology
6.
Exp Eye Res ; 212: 108767, 2021 11.
Article in English | MEDLINE | ID: mdl-34534542

ABSTRACT

Limbal stem cells (LSCs) are the stem cell reservoir for corneal epithelium. The protocol to isolate LSCs from human cornea has been examined and optimized. However, the isolation protocol has not been optimized for mouse cornea, which is crucial for the downstream cell analysis. Here we compared four different isolation methods evolved from the previous reports to obtain mouse limbal epithelial cells which are heterogeneous and contain LSCs in a single-cell suspension: (1) the dissected limbal rim was cut into pieces and digested by 10-cycle incubation in trypsin; (2) after the removal of corneal epithelium by a rotating bur, the remaining eyeball was incubated in dispase at 4 °C for overnight to obtain limbal epithelial sheet, followed by trypsin digestion into a single-cell suspension; (3) same as method 2 except that the incubation was in dispase at 37 °C for 2h and an additional collagenase incubation at 37 °C for 20 min; (4) same as method 3 except that the corneal epithelium was punctured by a 1.5 mm trephine instead of being removed by a rotating bur. Method 1 showed the lowest cell yield, the lowest percentage of single cells, and the lowest number of limbal epithelial stem/progenitor cells in the harvested cells among the four methods, thus not a recommended protocol. Method 2, 3, and 4 isolated a comparable number of K14+ and p63α-bright stem/progenitor cells per eye. The remaining eye globe after cell collection in the three methods showed a complete removal of limbal epithelium albeit different extent of corneal and limbal stromal digestion. Among the three methods, method 2 showed a higher cell viability than method 4; method 3 yielded the lowest cell number; method 4 led to the highest percentage of single cells in cell suspension. Results suggest that method 2, 3, and 4 are preferred methods to isolate heterogeneous-LSCs from mouse corneas.


Subject(s)
Epithelium, Corneal/cytology , Limbus Corneae/cytology , Stem Cells/cytology , Animals , Cell Count , Cell Separation , Cells, Cultured , Female , Male , Mice , Mice, Inbred C57BL , Models, Animal
7.
Exp Eye Res ; 203: 108400, 2021 02.
Article in English | MEDLINE | ID: mdl-33347868

ABSTRACT

Current research on healthy corneal stromal cells will typically use primary cells as they are the most representative of in vivo behaviour. Primary cells are normally isolated from the limbus of discarded donor peripheral corneal tissue left over from transplantation (due to its relative abundance). Therefore, the central part of the cornea is less used in research as this tissue is usually used for transplantation. In some cases, although rare, the whole cornea, can become available for research. It is important to keep in mind that these corneas often have longer storage time, but the use of the central tissue for research is even more interesting, as knowing what cells are being transplanted into recipients would be highly relevant. To this end, stromal cells were extracted from both the limbus and central button of healthy corneas donated for research. This allowed for important comparison between central and limbal cells in culture. Of interest here was the extraction method of stromal cells from the donor tissue. The two most common methods of extraction are enzyme digestion and explant migration. However, no work has been done to understand how each method relatively affects the extracted cells. The extraction method and location from which stromal cells are harvested seems to have a significant effect on the cell adherence, survival, and gene expression of the stromal cells in culture. Enzyme digested cells showed that limbal and central cells had different gene expressions prior to culture, with gene such as ALDH3A1 being much more expressed in limbal cells. Enzyme digesting the limbal ring seems to yield the hardiest populations of stromal cells, a desirable trait in the culture of primary cells.


Subject(s)
Cell Separation/methods , Corneal Keratocytes/physiology , Corneal Stroma/cytology , Limbus Corneae/cytology , Cell Culture Techniques , Cell Survival/physiology , Culture Media, Serum-Free , Cytoskeletal Proteins/genetics , Gene Expression Regulation/physiology , Humans , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Tissue Donors
8.
Exp Eye Res ; 205: 108526, 2021 04.
Article in English | MEDLINE | ID: mdl-33662355

ABSTRACT

Limbal Stem Cell Deficiency (LSCD), caused due to corneal injury, primarily by chemical/alkali burns, leads to compromised vision. Recently, several animal models of corneal alkali burn injury have become available. The majority of the studies with these animal models start interventions soon after the injury. However, in the clinical setting, there is a considerable delay before the intervention is initiated. Detailed knowledge of the molecular, histopathological, and clinical parameters associated with the progression of the injury leading to LSCD is highly desirable. In this context, we set out to investigate clinical, histopathological parameters of ocular surface alkali burn over a long period of time, post-injury. Limbal stem cell-deficient animal models of rabbits were created by alkali burn using sodium hydroxide, which was then assessed for their progression towards LSCD by grading the alkali burn, corneal haze, and vascularization. Additionally, cells present on the corneal surface after the burn was investigated by histology and immunophenotyping. Grading of rabbit eyes post-alkali burn had shown complete conjunctivalization in 80% (n = 12/15) of the rabbits with the alkali burn grade score of 3.88 ± 0.29 in three months and remained stable at four months (4.12 ± 0.24). However, ocular surface showed self-healing in 20% (n = 3/15) of the rabbits with a score of 1.67 ± 0.34 in four months irrespective of similar alkali injury. These self-healing corneas exhibited decreased opacity score from 2.51 ± 0.39 to 0.66 ± 0.22 (p = 0.002) and regressed vascularity from 1.66 ± 0.41 to 0.66 ± 0.33 in one to nine months, respectively. Restoration of the corneal phenotype (CK3+) was observed in central and mid-peripheral regions of the self-healing corneas, and histology revealed the localization of inflammatory cells to the peripheral cornea when compared to conjunctivalized and scarred LSCD eyes. Our study shows the essentiality to consider the time required for surgical intervention after the corneal alkali injury in rabbit models as evident from their tendency to self-heal and restore corneal phenotype without therapy. Such information on the possibility of self-healing should be useful in further studies as well as determining interventional timings and strategy during clinical presentation of corneal alkali burns.


Subject(s)
Burns, Chemical/physiopathology , Corneal Injuries/physiopathology , Corneal Neovascularization/physiopathology , Corneal Opacity/physiopathology , Eye Burns/chemically induced , Recovery of Function/physiology , Sodium Hydroxide/toxicity , Animals , Caustics/toxicity , Conjunctiva/physiopathology , Cornea/physiopathology , Disease Models, Animal , Eye Burns/physiopathology , Follow-Up Studies , Limbus Corneae/cytology , Rabbits , Stem Cell Transplantation , Wound Healing/physiology
9.
Exp Eye Res ; 211: 108720, 2021 10.
Article in English | MEDLINE | ID: mdl-34389315

ABSTRACT

The transplantation of expansions of limbal epithelial stem cells (LESC) remains one of the most efficient therapies for the treatment of limbal stem cell deficiency (LSCD) to date. However, the available donor corneas are scarce, and the corneas conserved for long time, under hypothermic conditions (after 7 days) or in culture (more than 28 days), are usually discarded due to poor viability of the endothelial cells. To establish an objective criterion for the utilisation or discarding of corneas as a source of LESC, we characterized, by immunohistochemistry analysis, donor corneas conserved in different conditions and for different periods of time. We also studied the potency of LESCs isolated from these corneas and maintained in culture up to 3 cell passages. We hoped that the study of markers of LESCs present in both the corneoscleral histological sections and the cell cultures would show the adequacy of the methods used for cell isolation and how fit the LESC enrichment of the obtained cell populations to be expanded was. Thus, the expressions of markers of the cells residing in the human limbal and corneal epithelium (cytokeratin CK15 and CK12, vimentin, Collagen VII, p63α, ABCG2, Ki67, Integrin ß4, ZO1, and melan A) were analysed in sections of corneoscleral tissues conserved in hypothermic conditions for 2-9 days with post-mortem time (pmt) < 8 h or for 1 day with pmt > 16 h, and in sclerocorneal rims maintained in an organ culture medium for 29 days. Cell populations isolated from donor corneoscleral tissues were also assessed based on these markers to verify the adequacy of isolation methods and the potential of expanding LESCs from these tissues. Positivity for several putative stem cell markers such as CK15 and p63α was detected in all corneoscleral tissues, although a decrease was recorded in the ones conserved for longer times. The barrier function and the ability to adhere to the extracellular matrix were maintained in all the analysed tissues. In limbal epithelial cell cultures, a simultaneous decrease in the melan A melanocyte marker and the putative stem cell markers was detected, suggesting a close relationship between the melanocytes and the limbal stem cells of the niche. Holoclones stained with putative stem cell markers were obtained from long-term, hypothermic, stored sclerocorneal rims. The results showed that the remaining sclerocorneal rims after corneal transplantation, which were conserved under hypothermic conditions for up to 7 days and would have been discarded at a first glance, still maintained their potential as a source of LESC cultures.


Subject(s)
Cornea/cytology , Epithelium, Corneal/cytology , Limbus Corneae/cytology , Organ Culture Techniques/methods , Stem Cells/cytology , Adult , Aged , Aged, 80 and over , Biomarkers/metabolism , Cell Separation , Cells, Cultured , Collagen/metabolism , Cornea/metabolism , Epithelium, Corneal/metabolism , Humans , Keratins/metabolism , Limbus Corneae/metabolism , Middle Aged , Stem Cells/metabolism , Time Factors , Tissue Donors , Tissue Preservation/methods , Vimentin/metabolism
10.
Ophthalmic Res ; 64(2): 145-167, 2021.
Article in English | MEDLINE | ID: mdl-32634808

ABSTRACT

To discuss and evaluate new technologies for a better diagnosis of corneal diseases and limbal stem cell deficiency, the outcomes of a consensus process within the European Vision Institute (and of a workshop at the University of Cologne) are outlined. Various technologies are presented and analyzed for their potential clinical use also in defining new end points in clinical trials. The disease areas which are discussed comprise dry eye and ocular surface inflammation, imaging, and corneal neovascularization and corneal grafting/stem cell and cell transplantation. The unmet needs in the abovementioned disease areas are discussed, and realistically achievable new technologies for better diagnosis and use in clinical trials are outlined. To sum up, it can be said that there are several new technologies that can improve current diagnostics in the field of ophthalmology in the near future and will have impact on clinical trial end point design.


Subject(s)
Clinical Trials as Topic , Corneal Diseases/surgery , Epithelium, Corneal/pathology , Limbus Corneae/cytology , Stem Cell Transplantation/methods , Stem Cells/cytology , Congresses as Topic , Corneal Diseases/metabolism , Corneal Diseases/pathology , Epithelium, Corneal/metabolism , Europe , Humans
11.
Int J Mol Sci ; 22(21)2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34769405

ABSTRACT

Limbal epithelial stem/progenitor cells (LSCs) reside in a niche that contains finely tuned balances of various signaling pathways including Wnt, Notch, BMP, Shh, YAP, and TGFß. The activation or inhibition of these pathways is frequently dependent on the interactions of LSCs with various niche cell types and extracellular substrates. In addition to receiving molecular signals from growth factors, cytokines, and other soluble molecules, LSCs also respond to their surrounding physical structure via mechanotransduction, interaction with the ECM, and interactions with other cell types. Damage to LSCs or their niche leads to limbal stem cell deficiency (LSCD). The field of LSCD treatment would greatly benefit from an understanding of the molecular regulation of LSCs in vitro and in vivo. This review synthesizes current literature around the niche factors and signaling pathways that influence LSC function. Future development of LSCD therapies should consider all these niche factors to achieve improved long-term restoration of the LSC population.


Subject(s)
Epithelium, Corneal/metabolism , Eye/physiopathology , Limbus Corneae/metabolism , Stem Cell Niche/physiology , Stem Cells/metabolism , Animals , Epithelium, Corneal/cytology , Eye/metabolism , Humans , Limbus Corneae/cytology , Mechanotransduction, Cellular/physiology , Stem Cells/cytology
12.
Int J Mol Sci ; 22(18)2021 Sep 17.
Article in English | MEDLINE | ID: mdl-34576227

ABSTRACT

The transplantation of ex vivo expanded limbal epithelial progenitor cells (LEPCs) on amniotic membrane or fibrin gel is an established therapeutic strategy to regenerate the damaged corneal surface in patients with limbal stem cell deficiency (LSCD), but the long-term success rate is restricted. A scaffold with niche-specific structure and extracellular matrix (ECM) composition might have the advantage to improve long-term clinical outcomes, in particular for patients with severe damage or complete loss of the limbal niche tissue structure. Therefore, we evaluated the decellularized human limbus (DHL) as a biomimetic scaffold for the transplantation of LEPCs. Corneoscleral tissue was decellularized by sodium deoxycholate and deoxyribonuclease I in the presence or absence of dextran. We evaluated the efficiency of decellularization and its effects on the ultrastructure and ECM composition of the human corneal limbus. The recellularization of these scaffolds was studied by plating cultured LEPCs and limbal melanocytes (LMs) or by allowing cells to migrate from the host tissue following a lamellar transplantation ex vivo. Our decellularization protocol rapidly and effectively removed cellular and nuclear material while preserving the native ECM composition. In vitro recellularization by LEPCs and LMs demonstrated the good biocompatibility of the DHL and intrastromal invasion of LEPCs. Ex vivo transplantation of DHL revealed complete epithelialization as well as melanocytic and stromal repopulation from the host tissue. Thus, the generated DHL scaffold could be a promising biological material as a carrier for the transplantation of LEPCs to treat LSCD.


Subject(s)
Corneal Diseases/metabolism , Limbus Corneae/cytology , Stem Cell Niche , Stem Cells/metabolism , Tissue Engineering/instrumentation , Adult , Aged , Aged, 80 and over , Amnion , Biomimetics , Cell Differentiation , Cell Transplantation/methods , Cells, Cultured , Dextrans/chemistry , Epithelial Cells/metabolism , Extracellular Matrix/metabolism , Humans , Melanocytes/metabolism , Middle Aged , Organ Culture Techniques , Phenotype , Tissue Engineering/methods
13.
Molecules ; 26(22)2021 Nov 11.
Article in English | MEDLINE | ID: mdl-34833901

ABSTRACT

A recombinant formulation of silk fibroin containing the arginine-glycine-aspartic acid (RGD) cell-binding motif (RGD-fibroin) offers potential advantages for the cultivation of corneal cells. Thus, we investigated the growth of corneal stromal cells and epithelial cells on surfaces created from RGD-fibroin, in comparison to the naturally occurring Bombyx mori silk fibroin. The attachment of cells was compared in the presence or absence of serum over a 90 min period and analyzed by quantification of dsDNA content. Stratification of epithelial cells on freestanding membranes was examined by confocal fluorescence microscopy and optimized through use of low molecular weight poly(ethylene glycol) (PEG; 300 Da) as a porogen, the enzyme horseradish peroxidase (HRP) as a crosslinking agent, and stromal cells grown on the opposing membrane surface. The RGD-fibroin reduced the tendency of stromal cell cultures to form clumps and encouraged the stratification of epithelial cells. PEG used in conjunction with HRP supported the fabrication of more permeable freestanding RGD-fibroin membranes, that provide an effective scaffold for stromal-epithelial co-cultures. Our studies encourage the use of RGD-fibroin for corneal cell culture. Further studies are required to confirm if the benefits of this formulation are due to changes in the expression of integrins, components of the extracellular matrix, or other events at the transcriptional level.


Subject(s)
Cornea/cytology , Fibroins/chemistry , Tissue Scaffolds/chemistry , Animals , Biomechanical Phenomena , Bombyx/chemistry , Bombyx/genetics , Cell Adhesion , Cell Proliferation , Cells, Cultured , Coculture Techniques , Corneal Stroma/cytology , Epithelium, Corneal/cytology , Fibroins/genetics , Humans , Limbus Corneae/cytology , Membranes, Artificial , Microscopy, Confocal , Oligopeptides/chemistry , Oligopeptides/genetics , Permeability , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Tissue Engineering
14.
J Cell Mol Med ; 24(20): 12020-12031, 2020 10.
Article in English | MEDLINE | ID: mdl-32914934

ABSTRACT

Homeostasis and function of limbal epithelial stem cells (LESCs) rely on the limbal niche, which, if dysfunctional, leads to limbal epithelial stem cell deficiency (LSCD) and impaired vision. Hence, recovery of niche function is a principal therapeutic goal in LSCD, but the molecular mechanisms of limbal niche homeostasis are still largely unknown. Here, we report that the neural crest transcription factor SOX10, which is expressed in neural crest-derived limbal niche cells (LNCs), is required for LNCs to promote survival of LESCs both in vivo and in vitro. In fact, using mice with a Sox10 mutation and in vitro coculture experiments, we show that SOX10 in LNCs stimulates the production of KIT ligand (KITL), which in turn activates in LESCs the KIT-AKT signalling pathway that protects the cells against activated CASPASE 3-associated cell death. These results suggest that SOX10 and the KITL/KIT-AKT pathway play key roles in limbal niche homeostasis and LESC survival. These findings provide molecular insights into limbal niche function and may point to rational approaches for therapeutic interventions in LSCD.


Subject(s)
Epithelial Cells/cytology , Limbus Corneae/cytology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-kit/metabolism , SOXE Transcription Factors/metabolism , Stem Cell Factor/metabolism , Stem Cell Niche , Stem Cells/metabolism , Animals , Cell Survival , Epithelial Cells/metabolism , Mice , Paracrine Communication , Signal Transduction
15.
Development ; 144(13): 2338-2351, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28559289

ABSTRACT

Corneal epithelial stem cells residing within the annular limbal crypts regulate adult tissue homeostasis. Autologous limbal grafts and tissue-engineered corneal epithelial cell sheets have been widely used in the treatment of various ocular surface defects. In the case of bilateral limbal defects, pluripotent stem cell (PSC)-derived corneal epithelial cells are now being explored as an alternative to allogeneic limbal grafts. Here, we report an efficient method to generate complex three-dimensional corneal organoids from human PSCs. The eye field primordial clusters that emerged from differentiating PSCs developed into whole eyeball-like, self-organized, three-dimensional, miniature structures consisting of retinal primordia, corneal primordia, a primitive eyelid-like outer covering and ciliary margin zone-like adnexal tissues in a stepwise maturation process within 15 weeks. These minicorneal organoids recapitulate the early developmental events in vitro and display similar anatomical features and marker expression profiles to adult corneal tissues. They offer an alternative tissue source for regenerating different layers of the cornea and eliminate the need for complicated cell enrichment procedures.


Subject(s)
Cornea/cytology , Induced Pluripotent Stem Cells/cytology , Organoids/cytology , Amnion/cytology , Biomarkers/metabolism , Cell Adhesion , Cell Differentiation , Conjunctiva/cytology , Corneal Transplantation , Epithelium, Corneal/cytology , Humans , Immunohistochemistry , Induced Pluripotent Stem Cells/metabolism , Limbus Corneae/cytology , Models, Biological
16.
Mol Vis ; 26: 434-444, 2020.
Article in English | MEDLINE | ID: mdl-32565671

ABSTRACT

Chronic elevation of intraocular pressure (IOP) is a major risk factor associated with primary open angle glaucoma (POAG), a common form of progressive optic neuropathy that can lead to debilitating loss of vision. Recent studies have identified the role of nitric oxide (NO) in the regulation of IOP, and as a result, several therapeutic ventures are currently targeting enhancement of NO signaling in the eye. Although a low level of NO is important for ocular physiology, excess exogenous NO can be detrimental. Therefore, the ability to directly measure NO in real time is essential for determining the role of NO signaling in glaucomatous pathophysiology. Historically, NO activity in human tissues has been determined by indirect methods that measure levels of NO metabolites (nitrate/nitrite) or downstream components of the NO signaling pathway (cGMP). In this proof-of-concept work, we assess the feasibility of direct, real-time measurement of NO in ex vivo cultured human corneoscleral segments using electrochemistry. A NO-selective electrode (ISO-NOPF200) paired to a free radical analyzer (TBR1025) was placed on the trabecular meshwork (TM) rim for real-time measurement of NO released from cells. Exogenous NO produced within cells was measured after treatment of corneoscleral segments with esterase-dependent NO-donor O2-acetoxymethylated diazeniumdiolate (DETA-NONOate/AM; 20 µM) and latanoprostene bunod (5-20 µM). A fluorescent NO-binding dye DAF-FM (4-Amino-5-methylamino- 2',7'-difluorofluorescein diacetate) was used for validation. A linear relationship was observed between the electric currents measured by the NO-sensing electrode and the NO standard concentrations, establishing a robust calibration curve. Treatment of ex vivo cultured human donor corneoscleral segments with DETA-NONOate/AM and latanoprostene bunod led to a significant increase in NO production compared with vehicle-treated controls, as detected electrochemically. Furthermore, the DAF-FM fluorescence intensity was higher in outflow pathway tissues of corneoscleral segments treated with DETA-NONOate/AM and latanoprostene bunod compared with vehicle-treated controls. In conclusion, these results demonstrate that NO-sensing electrodes can be used to directly measure NO levels in real time from the tissues of the outflow pathway.


Subject(s)
Electrochemistry/instrumentation , Electrochemistry/methods , Limbus Corneae/metabolism , Nitric Oxide/metabolism , Ocular Hypertension/metabolism , Tonometry, Ocular/instrumentation , Tonometry, Ocular/methods , Azo Compounds/chemistry , Cells, Cultured , Electrodes , Fluoresceins/chemistry , Humans , Intraocular Pressure , Limbus Corneae/cytology , Prostaglandins F, Synthetic/chemistry , Trabecular Meshwork/metabolism
17.
Nature ; 511(7509): 353-7, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25030174

ABSTRACT

Corneal epithelial homeostasis and regeneration are sustained by limbal stem cells (LSCs), and LSC deficiency is a major cause of blindness worldwide. Transplantation is often the only therapeutic option available to patients with LSC deficiency. However, while transplant success depends foremost on LSC frequency within grafts, a gene allowing for prospective LSC enrichment has not been identified so far. Here we show that ATP-binding cassette, sub-family B, member 5 (ABCB5) marks LSCs and is required for LSC maintenance, corneal development and repair. Furthermore, we demonstrate that prospectively isolated human or murine ABCB5-positive LSCs possess the exclusive capacity to fully restore the cornea upon grafting to LSC-deficient mice in xenogeneic or syngeneic transplantation models. ABCB5 is preferentially expressed on label-retaining LSCs in mice and p63α-positive LSCs in humans. Consistent with these findings, ABCB5-positive LSC frequency is reduced in LSC-deficient patients. Abcb5 loss of function in Abcb5 knockout mice causes depletion of quiescent LSCs due to enhanced proliferation and apoptosis, and results in defective corneal differentiation and wound healing. Our results from gene knockout studies, LSC tracing and transplantation models, as well as phenotypic and functional analyses of human biopsy specimens, provide converging lines of evidence that ABCB5 identifies mammalian LSCs. Identification and prospective isolation of molecularly defined LSCs with essential functions in corneal development and repair has important implications for the treatment of corneal disease, particularly corneal blindness due to LSC deficiency.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP-Binding Cassette Transporters/metabolism , Limbus Corneae/cytology , Limbus Corneae/physiology , Regeneration , Stem Cells/metabolism , Wound Healing , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/deficiency , ATP-Binding Cassette Transporters/deficiency , Animals , Apoptosis , Biomarkers/metabolism , Cell Differentiation , Cell Proliferation , Female , Humans , Male , Mice , Mice, Knockout , Molecular Sequence Data , Stem Cell Transplantation , Stem Cells/cytology , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
18.
Nature ; 511(7509): 358-61, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25030175

ABSTRACT

The surface of the cornea consists of a unique type of non-keratinized epithelial cells arranged in an orderly fashion, and this is essential for vision by maintaining transparency for light transmission. Cornea epithelial cells (CECs) undergo continuous renewal from limbal stem or progenitor cells (LSCs), and deficiency in LSCs or corneal epithelium--which turns cornea into a non-transparent, keratinized skin-like epithelium--causes corneal surface disease that leads to blindness in millions of people worldwide. How LSCs are maintained and differentiated into corneal epithelium in healthy individuals and which key molecular events are defective in patients have been largely unknown. Here we report establishment of an in vitro feeder-cell-free LSC expansion and three-dimensional corneal differentiation protocol in which we found that the transcription factors p63 (tumour protein 63) and PAX6 (paired box protein PAX6) act together to specify LSCs, and WNT7A controls corneal epithelium differentiation through PAX6. Loss of WNT7A or PAX6 induces LSCs into skin-like epithelium, a critical defect tightly linked to common human corneal diseases. Notably, transduction of PAX6 in skin epithelial stem cells is sufficient to convert them to LSC-like cells, and upon transplantation onto eyes in a rabbit corneal injury model, these reprogrammed cells are able to replenish CECs and repair damaged corneal surface. These findings suggest a central role of the WNT7A-PAX6 axis in corneal epithelial cell fate determination, and point to a new strategy for treating corneal surface diseases.


Subject(s)
Corneal Diseases/metabolism , Corneal Diseases/pathology , Epithelium, Corneal/cytology , Epithelium, Corneal/metabolism , Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Homeostasis , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , Wnt Proteins/metabolism , Animals , Cell Differentiation , Cell Lineage , Disease Models, Animal , Epithelium, Corneal/pathology , Eye Proteins/genetics , Homeodomain Proteins/genetics , Humans , Limbus Corneae/cytology , Limbus Corneae/metabolism , Male , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Rabbits , Repressor Proteins/genetics , Signal Transduction , Skin/cytology , Skin/metabolism , Skin/pathology , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Wnt Proteins/genetics
19.
Int J Med Sci ; 17(4): 549-557, 2020.
Article in English | MEDLINE | ID: mdl-32174786

ABSTRACT

Purpose: To compare the difference in gene expression between human limbal niche cells (LNC) and bone marrow derived mesenchymal stem cells (BMMSC). Methods: LNC were isolated by collagenase and expanded in modified embryonic stem cell medium (MESCM) on a Matrigel coated plastic plate. Cell diameters were measured with Image J software. Relative gene expression levels between LNC and BMMSC were compared using Affymetrix Human Primer View Gene Expression Array. A subset of differentially expressed genes was verified by RT-qPCR. The protein level of LAMA1 and COL4A1 was confirmed by Western blot and immunostaining. Results: The average diameter of LNC was 10.2±2.4 µm, which was significantly smaller than that of BMMSC (14 ±3.4 µm) (p<0.0001). Expression of 20,432 genes was examined by Gene Expression Array, among which expression of 349 genes in LNC was 10-fold or higher than that of BMMSC and expression of 8 genes in LNC was 100-fold or higher than that of BMMSC, while expression of 3 genes in BMMSC was 100-fold higher than that of LNC. GO analysis and pathway analysis showed that the differentially expressed genes were mainly enriched in the extracellular matrix receptor interaction pathway and Wnt signaling pathway. In addition, RT-qPCR results demonstrated that the expression of CD73, CD90, CD105, PDGFRß, Vimentin, SCF, KIT (CD117), COL14A1, LAMA2, THBS2, FZD1, BMP2 and CXCL12 genes in LNC were at least 2 folds higher than BMMSC. The protein level of LAMA1 was higher but the protein level of COL4A1 was lower in LNC than that in BMMSC. Conclusion: LNC exhibit differential gene expression from BMMSC in the extracellular matrix (ECM) receptor interaction pathway and Wnt signaling pathway, suggesting that LNC have their unique signaling pathways to support limbal stem cell niches.


Subject(s)
Bone Marrow Cells/cytology , Limbus Corneae/cytology , Mesenchymal Stem Cells/cytology , Stem Cell Niche , Cell Differentiation , Cells, Cultured , Collagen Type IV/metabolism , Culture Media , Gene Expression Profiling , Gene Expression Regulation , Humans , Laminin/metabolism , Oligonucleotide Array Sequence Analysis , Wnt Signaling Pathway
20.
BMC Ophthalmol ; 20(1): 120, 2020 Mar 26.
Article in English | MEDLINE | ID: mdl-32216768

ABSTRACT

BACKGROUND: The cellular process of autophagy is essential for maintaining the health of ocular tissue. Dysregulation of autophagy is associated with several ocular diseases including keratoconus and macular degeneration. It is known that autophagy can be used to respond to microbial infections and that certain microbes can exploit the autophagic process to their benefit. In this study, a genetic approach was used to identify surface-associated and secreted products generated by the opportunistic pathogen Serratia marcescens involved in activation of autophagy. METHODS: A recombinant human corneal limbal epithelial cell line expressing a LC3-GFP fusion protein was challenged with normalized secretomes from wild-type and mutant S. marcescens derivatives. LC3-GFP fluorescence patterns were used to assess the ability of wild-type and mutant bacteria to influence autophagy. Purified prodigiosin was obtained from stationary phase bacteria and used to challenge ocular cells. RESULTS: Mutations in the global regulators eepR and gumB genes highly reduced the ability of the bacteria to activate autophagy in corneal cells. This effect was further narrowed down to the secreted cytolysin ShlA and the biologically active pigment prodigiosin. Purified prodigiosin and ShlA from Escherichia coli further supported the role of these factors in activating autophagy in human corneal cells. Additional genetic data indicate a role for flagellin and type I pili, but not the nuclease, S-layer protein, or serratamolide biosurfactant in activation of autophagy. CONCLUSIONS: This work identifies specific bacterial components that activate autophagy and give insight into potential host-pathogen interactions or compounds that can be used to therapeutically manipulate autophagy.


Subject(s)
Autophagy/drug effects , Bacterial Proteins/pharmacology , Epithelium, Corneal/pathology , Hemolysin Proteins/pharmacology , Limbus Corneae/cytology , Prodigiosin/pharmacology , Serratia marcescens/pathogenicity , Adenine/analogs & derivatives , Adenine/pharmacology , Bacterial Physiological Phenomena , Cell Line , Green Fluorescent Proteins , Humans , Keratitis/microbiology , Microscopy, Confocal , Perforin , Serratia marcescens/genetics , Serratia marcescens/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL