Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Pathol ; 188(9): 2016-2024, 2018 09.
Article in English | MEDLINE | ID: mdl-30097165

ABSTRACT

Corneal transplantation is the most prevalent form of tissue transplantation. The success of corneal transplantation mainly relies on the integrity of corneal endothelial cells (CEnCs), which maintain graft transparency. CEnC density decreases significantly after corneal transplantation even in the absence of graft rejection. To date, different strategies have been used to enhance CEnC survival. The neuropeptide vasoactive intestinal peptide (VIP) improves CEnC integrity during donor cornea tissue storage and protects CEnCs against oxidative stress-induced apoptosis. However, little is known about the effect of exogenous administration of VIP on corneal transplant outcomes. We found that VIP significantly accelerates endothelial wound closure and suppresses interferon-γ- and tumor necrosis factor-α-induced CEnC apoptosis in vitro in a dose-dependent manner. In addition, we found that intracameral administration of VIP to mice undergoing syngeneic corneal transplantation with endothelial injury increases CEnC density and decreases graft opacity scores. Finally, using a mouse model of allogeneic corneal transplantation, we found for the first time that treatment with VIP significantly suppresses posttransplantation CEnC loss and improves corneal allograft survival.


Subject(s)
Corneal Transplantation , Endothelium, Corneal/drug effects , Gastrointestinal Agents/pharmacology , Graft Rejection/prevention & control , Graft Survival/drug effects , Vasoactive Intestinal Peptide/pharmacology , Wound Healing/drug effects , Allografts , Animals , Cells, Cultured , Endothelium, Corneal/injuries , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
2.
Mol Ther ; 25(8): 1881-1888, 2017 08 02.
Article in English | MEDLINE | ID: mdl-28502469

ABSTRACT

Corneal injuries are among the major causes of ocular morbidity and vision impairment. Optimal epithelial wound healing is critical for the integrity and transparency of the cornea after injury. Hepatocyte growth factor (HGF) is a mitogen and motility factor that primarily regulates epithelial cell function. Herein, we investigate the effect of HGF on proliferation of corneal epithelial cells (CECs) in inflamed conditions both in vitro and in vivo. We demonstrate that HGF not only promotes CEC proliferation in homeostatic conditions but also reverses the anti-proliferative effect of the inflammatory environment on these cells. Furthermore, using a mouse model of ocular injury, we show that HGF treatment suppresses ocular inflammation and actively augments CEC proliferation, leading to improved and accelerated corneal epithelial repair. These findings have potential translational implications and could provide a framework for the development of novel HGF-based therapies for corneal epithelial defects.


Subject(s)
Corneal Injuries/genetics , Corneal Injuries/metabolism , Epithelium/metabolism , Hepatocyte Growth Factor/genetics , Wound Healing/genetics , Animals , Biomarkers , CD11b Antigen/metabolism , Cell Proliferation , Corneal Injuries/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Epithelium/pathology , Gene Expression , Hepatocyte Growth Factor/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/therapy , Inflammation Mediators/metabolism , Mice
3.
Stem Cell Reports ; 7(4): 583-590, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27693426

ABSTRACT

Transparency of the cornea is indispensable for optimal vision. Ocular trauma is a leading cause of corneal opacity, leading to 25 million cases of blindness annually. Recently, mesenchymal stem cells (MSCs) have gained prominence due to their inflammation-suppressing and tissue repair functions. Here, we investigate the potential of MSCs to restore corneal transparency following ocular injury. Using an in vivo mouse model of ocular injury, we report that MSCs have the capacity to restore corneal transparency by secreting high levels of hepatocyte growth factor (HGF). Interestingly, our data also show that HGF alone can restore corneal transparency, an observation that has translational implications for the development of HGF-based therapy.


Subject(s)
Cornea/physiology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Regeneration , Animals , Cell Line , Cornea/cytology , Cornea/drug effects , Corneal Injuries/genetics , Corneal Injuries/metabolism , Corneal Injuries/pathology , Corneal Injuries/therapy , Fibroblasts , Gene Expression , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/pharmacology , Humans , Interleukin-1beta/pharmacology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Mice , Models, Animal , RNA, Small Interfering/genetics , Wound Healing
4.
Int Rev Cell Mol Biol ; 319: 45-106, 2015.
Article in English | MEDLINE | ID: mdl-26404466

ABSTRACT

The potential cause of blindness worldwide includes diseases of the cornea, ocular surface (limbal stem cell deficiency, allergic conjunctivitis, dry eye diseases), and retinal diseases. The presence of stem cells (limbal stem cells) in the basal region of the limbus makes it an important tool for the ocular regeneration and also in maintaining the transparency of eye by replacing the corneal epithelium continuously. Various surgical modalities have been developed like cultured limbal epithelial transplantation, cultured oral mucosal epithelial transplantation, simple limbal epithelial transplantation, etc., utilizing the cell-based regenerative properties to treat limbal disorder. Cell-based therapies for ocular repair and regeneration comprise a major hope by therapies involving the mesenchymal stem cells, embryonic stem cells, and limbal stem cells for the restoration of vision in individuals whose ocular tissue has been irreversibly damaged by disease or trauma. This review explores critical needs in human disease mainly the ocular problem where cell-based therapeutics is exceptionally well suited and also the use of animal models, various artificial scaffolds, as well as advancement in clinical technique to challenge the current demand to overcome corneal blindness.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Ocular Physiological Phenomena , Regeneration/physiology , Animals , Epithelium, Corneal/cytology , Epithelium, Corneal/pathology , Epithelium, Corneal/physiology , Eye Diseases/pathology , Eye Diseases/physiopathology , Humans , Stem Cells/cytology , Stem Cells/physiology
5.
Invest Ophthalmol Vis Sci ; 55(10): 6631-8, 2014 Sep 16.
Article in English | MEDLINE | ID: mdl-25228546

ABSTRACT

PURPOSE: To investigate whether systemically injected syngeneic mesenchymal stem cells (MSCs) can home to the transplanted cornea, suppress induction of alloimmunity, and promote allograft survival. METHODS: Mesenchymal stem cells were generated from bone marrow of wild-type BALB/c or GFP (green fluorescent protein)+ C57BL/6 mice, and 1×10(6) cells were intravenously injected to allografted recipients 3 hours after surgery. Mesenchymal stem cells homing to the cornea were examined at day 3 post transplantation by immunohistochemistry. MHC (major histocompatibility complex) II+CD11c+ cells were detected in the cornea and lymph nodes (LNs) 14 days post transplantation using flow cytometry. Cytokine expression of bone marrow-derived dendritic cells (BMDCs) was determined using real-time PCR. ELISPOT assay was used to assess indirect and direct host T cell allosensitization, and graft survival was evaluated by slit-lamp biomicroscopy weekly up to 8 weeks. RESULTS: Intravenously injected GFP+ MSCs were found in abundance in the transplanted cornea, conjunctiva, and LNs, but not in the ungrafted (contralateral) tissue. The frequencies of mature MHC II+CD11c+ antigen-presenting cells (APCs) were substantially decreased in the corneas and draining LNs of MSC-injected allograft recipients compared to control recipients. Maturation and function of in vitro cultured BMDCs were decreased when cocultured with MSCs. Draining LNs of MSC-injected allograft recipients showed lower frequencies of IFNγ-secreting Th1 cells compared to the control group. Allograft survival rate was significantly higher in MSC-injected recipients compared to non-MSC-injected recipients. CONCLUSIONS: Our data demonstrate that systemically administered MSCs specifically home to the inflamed ocular surface and promote allograft survival by inhibiting APC maturation and induction of alloreactive T cells.


Subject(s)
Corneal Transplantation/methods , Dendritic Cells/immunology , Graft Rejection/immunology , Major Histocompatibility Complex/immunology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/immunology , Th1 Cells/immunology , Animals , Autoimmunity , Cells, Cultured , Cytokines/biosynthesis , Cytokines/genetics , Disease Models, Animal , Eye Burns/chemically induced , Eye Burns/pathology , Eye Burns/surgery , Flow Cytometry , Gene Expression Regulation , Graft Rejection/pathology , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Fluorescence , RNA/genetics , Real-Time Polymerase Chain Reaction , Transplantation, Homologous
6.
Invest Ophthalmol Vis Sci ; 55(9): 5795-805, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25125605

ABSTRACT

PURPOSE: The improved surgical outcomes associated with transplantation of cultivated amniotic membrane expanded limbal epithelium (AMLE) compared to traditional donor methods has led to substantial adoption of this technique for treatment of limbal stem cell deficiency. METHODS: The mRNA expression profiles of AMLE and CE were assayed using microarrays. Transcripts with a 1.5-fold change in either direction in addition to a Bonferroni adjusted P value < 0.05 were considered to be differentially expressed. Expression changes detected by microarray profiling and important corneal-limbal markers were assessed using quantitative real-time PCR (qRT-PCR) and immunofluorescence staining. RESULTS: A total of 487 probe sets (319 upregulated and 168 downregulated) were found to be differentially expressed between AMLE and CE. Enrichment analysis revealed significant overrepresentation of multiple biological processes (e.g., response to wounding, wound healing, and regulation of cell morphogenesis) within the differentially expressed gene list. The expression of a number of genes that were upregulated (ABCG2, S100A9, ITGA5, TIMP2, FGF5, PDGFC, SEMA3A) and downregulated (KLF4, P63α) in AMLE was confirmed using qRT-PCR. Immunofluorescence confirmed that AMLE cultures were P63α, ABCG2, CK3, CK12, and E-cadherin (E-cad) positive. CONCLUSIONS: In this study, we have shown that genes associated with wound healing processes are upregulated in AMLE. These gene expression changes may contribute to corneal restoration and the positive outcomes associated with transplantation.


Subject(s)
Epithelium, Corneal/metabolism , Limbus Corneae/metabolism , Wound Healing/physiology , Amnion/cytology , Biomarkers/metabolism , Cadaver , Cells, Cultured , Corneal Transplantation , Gene Expression Profiling , Humans , Kruppel-Like Factor 4 , Microarray Analysis , RNA/metabolism , RNA, Mitochondrial
7.
Invest Ophthalmol Vis Sci ; 53(2): 867-72, 2012 Feb 21.
Article in English | MEDLINE | ID: mdl-22232434

ABSTRACT

PURPOSE: Neurotrophic keratopathy (NK) is a corneal degeneration associated with corneal nerve dysfunction. It can cause corneal epithelial defects, stromal thinning, and perforation. However, it is not clear if and to which extent epithelial stem cells are affected in NK. The purpose of this study was to identify the relationship between corneolimbal epithelial progenitor/stem cells and sensory nerves using a denervated mouse model of NK. METHODS: NK was induced in mice by electrocoagulation of the ophthalmic branch of the trigeminal nerve. The absence of corneal nerves was confirmed with ß-III tubulin immunostaining and blink reflex test after 7 days. ATP-binding cassette subfamily G member 2 (ABCG2), p63, and hairy enhancer of split 1 (Hes1) were chosen as corneolimbal stem/progenitor cell markers and assessed in denervated mice versus controls by immunofluorescent microscopy and real-time PCR. In addition, corneolimbal stem/progenitor cells were detected as side population cells using flow cytometry, and colony-forming efficiency assay was performed to assess their function. RESULTS: ABCG2, p63, and Hes1 immunostaining were significantly decreased in denervated eyes after 7 days. Similarly, the expression levels of ABCG2, p63, K15, Hes1, and N-cadherin transcripts were also significantly decreased in denervated eyes. Stem/progenitor cells measured as side population from NK mice were decreased by approximately 75% compared with normals. In addition, the authors found a significant (P = 0.038) reduction in colony-forming efficiency of stem/progenitor cells harvested from denervated eyes. CONCLUSIONS: Corneolimbal stem/progenitor cells are significantly reduced after depletion of sensory nerves. The data suggest a critical role of innervation in maintaining stem cells and/or the stem cell niche.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Cornea/innervation , Corneal Diseases/pathology , Gene Expression Regulation , Homeodomain Proteins/genetics , Phosphoproteins/genetics , Stem Cells/pathology , Trans-Activators/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/biosynthesis , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Biomarkers/metabolism , Cell Survival , Cornea/metabolism , Cornea/pathology , Corneal Diseases/etiology , Corneal Diseases/genetics , Denervation/methods , Disease Models, Animal , Electrolysis , Epithelium, Corneal/metabolism , Epithelium, Corneal/pathology , Flow Cytometry , Genes, Tumor Suppressor , Helix-Loop-Helix Motifs , Homeodomain Proteins/biosynthesis , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Phosphoproteins/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Stem Cells/metabolism , Trans-Activators/biosynthesis , Transcription Factor HES-1 , Trigeminal Nerve Injuries/complications , Trigeminal Nerve Injuries/metabolism , Trigeminal Nerve Injuries/pathology
8.
Stem Cell Rev Rep ; 8(3): 696-705, 2012 Sep.
Article in English | MEDLINE | ID: mdl-21964568

ABSTRACT

Corneal transplantation with cultivated limbal or oral epithelium is a feasible treatment option for limbal stem cell deficiency (LSCD). Currently utilized co-culture of stem cells with murine 3T3 feeder layer renders the epithelial constructs as xenografts. To overcome the potential risks involved with xenotransplantation, we investigated the use of human-derived feeder layers for the ex vivo expansion of epithelial (stem) cells. Human limbal and oral epithelium was co-cultured with mouse 3T3 fibroblasts, human dermal fibroblasts (DF), human mesenchymal stem cells (MSC), and with no feeder cells (NF). Cell morphology was monitored with phase-contrast microscopy, and stem cell characteristics were assessed by immunohistochemistry, real-time PCR for p63 and ABCG2, (stem cell markers), and by colony-forming efficiency (CFE) assay. Immunohistochemical analysis detected positive staining for CK3 (cornea specific marker) and Iß1 and p63 (putative stem cell markers) in all culture conditions. The level of Iß1 and p63 was significantly higher in both limbal and oral cells cultured on the 3T3 feeder, as compared to the MSC or NF group (p<0.01). This level was comparable to the cells cultured on DF. Expression of p63 and ABCG2 in limbal and oral epithelial cells in the 3T3 and DF groups was significantly higher than that in the MSC or NF group (p<0.01). No statistical difference was detected between 3T3 and DF groups. The CFE of both limbal and oral cells co-cultured on 3T3 fibroblasts was comparable to cells grown on DF, and was significantly higher than that of cells co-cultured with MSC or NF (p<0.01). Epithelial cells grown on a DF feeder layer maintained a stem cell-like phenotype, comparable to cells grown on a 3T3 feeder layer. In conclusion, DF provides a promising substitute for 3T3 feeder cells during cultivation of xenobiotic-free corneal equivalents.


Subject(s)
Epithelium, Corneal/physiology , Feeder Cells/physiology , Fibroblasts/physiology , Limbus Corneae/cytology , Mouth Mucosa/cytology , 3T3 Cells , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Amnion/cytology , Animals , Cell Proliferation , Cell Shape , Coculture Techniques , Colony-Forming Units Assay , Dermis/cytology , Epithelium, Corneal/metabolism , Eye Diseases/therapy , Humans , Keratin-3/metabolism , Membrane Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Mice , Neoplasm Proteins/metabolism , Tissue Culture Techniques
SELECTION OF CITATIONS
SEARCH DETAIL