Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Proc Natl Acad Sci U S A ; 120(13): e2217576120, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36943878

ABSTRACT

Diabetes can result in impaired corneal wound healing. Mitochondrial dysfunction plays an important role in diabetic complications. However, the regulation of mitochondria function in the diabetic cornea and its impacts on wound healing remain elusive. The present study aimed to explore the molecular basis for the disturbed mitochondrial metabolism and subsequent wound healing impairment in the diabetic cornea. Seahorse analysis showed that mitochondrial oxidative phosphorylation is a major source of ATP production in human corneal epithelial cells. Live corneal biopsy punches from type 1 and type 2 diabetic mouse models showed impaired mitochondrial functions, correlating with impaired corneal wound healing, compared to nondiabetic controls. To approach the molecular basis for the impaired mitochondrial function, we found that Peroxisome Proliferator-Activated Receptor-α (PPARα) expression was downregulated in diabetic human corneas. Even without diabetes, global PPARα knockout mice and corneal epithelium-specific PPARα conditional knockout mice showed disturbed mitochondrial function and delayed wound healing in the cornea, similar to that in diabetic corneas. In contrast, fenofibrate, a PPARα agonist, ameliorated mitochondrial dysfunction and enhanced wound healing in the corneas of diabetic mice. Similarly, corneal epithelium-specific PPARα transgenic overexpression improved mitochondrial function and enhanced wound healing in the cornea. Furthermore, PPARα agonist ameliorated the mitochondrial dysfunction in primary human corneal epithelial cells exposed to diabetic stressors, which was impeded by siRNA knockdown of PPARα, suggesting a PPARα-dependent mechanism. These findings suggest that downregulation of PPARα plays an important role in the impaired mitochondrial function in the corneal epithelium and delayed corneal wound healing in diabetes.


Subject(s)
Diabetes Mellitus, Experimental , PPAR alpha , Mice , Humans , Animals , PPAR alpha/genetics , PPAR alpha/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Cornea/metabolism , Wound Healing/physiology , Mice, Knockout , Mitochondria/metabolism
2.
Anal Cell Pathol (Amst) ; 2022: 6718566, 2022.
Article in English | MEDLINE | ID: mdl-36340268

ABSTRACT

Diabetes mellitus (DM) is a group of metabolic diseases that is known to cause structural and functional ocular complications. In the human cornea, DM-related complications affect the epithelium, stroma, and nerves. Monocarboxylate transporters (MCTs) are a family of proton-linked plasma membrane transporters that carry monocarboxylates across plasma membranes. In the context of corneal health and disease, their role, presence, and function are largely undetermined and solely focused on the most common MCT isoforms, 1 through 4. In this study, we investigated the regulation of MCT1, 2, 4, 5, 8, and 10, in corneal DM, using established 3D self-assembled extracellular matrix (ECM) in vitro models. Primary stromal corneal fibroblasts were isolated from healthy (HCFs), type I (T1DMs), and type II (T2DMs) DM donors. Monoculture 3D constructs were created by stimulating stromal cells on transwells with stable vitamin C for two or four weeks. Coculture 3D constructs were created by adding SH-SY5Y neurons at two different densities, 12 k and 500 k, on top of the monocultures. Our data showed significant upregulation of MCT1 at 4 weeks for HCF, T1DM, and T2DM monocultures, as well as the 500 k nerve cocultures. MCT8 was significantly upregulated in HCF and T1DM monocultures and all of the 500 k nerve cocultures. Further, MCT10 was only expressed at 4 weeks for all cocultures and was limited to HCFs and T1DMs in monocultures. Immunofluorescence analysis showed cytoplasmic MCT expression for all cell types and significant downregulation of both MCT2 and MCT4 in HCFs, when compared to T1DMs and T2DMs. Herein, we reveal the existence and modulation of MCTs in the human diabetic cornea in vitro. Changes appeared dependent on neuronal density, suggesting that MCTs are very likely critical to the neuronal defects observed in diabetic keratopathy/neuropathy. Further studies are warranted in order to fully delineate the role of MCTs in corneal diabetes.


Subject(s)
Diabetes Mellitus, Type 1 , Neuroblastoma , Humans , Monocarboxylic Acid Transporters/metabolism , Cornea/metabolism , Protein Isoforms/metabolism
3.
Bioengineered ; 13(5): 12955-12971, 2022 05.
Article in English | MEDLINE | ID: mdl-35615975

ABSTRACT

Etiology and pathogenesis of age-related cataract is not entirely clear till now. Senescence marker protein 30 (SMP30) is a newly discovered anti-aging factor, which plays an important role in preventing apoptosis and reducing oxidative stress damage. Mitochondria are located at the intersection of key cellular pathways, such as energy substrate metabolism, reactive oxygen species (ROS) production and apoptosis. Oxidative stress induced by 4-hydroxynonenal (4-HNE) is closely related to neurodegenerative diseases and aging. Our study focused on the effect of SMP30 on mitochondrial homeostasis of human lens epithelial cells (HLECs) induced by 4-HNE. Western blots and qPCR were used to compare the expression of SMP30 protein in the residual lens epithelial cells in the lens capsule of age-related cataract (ARC) patients and the donated transparent lens capsule. On this basis, SMP30 overexpression plasmid and SMP30 shRNA interference plasmid were introduced to explore the effect of SMP30 on the biological behavior in HLECs under the condition of oxidative stress induced by 4-HNE through immunohistochemistry, ROS evaluation, metabolic spectrum analysis and JC-1 fluorescence measurement. Given that Nuclear Factor erythroid 2-Related Factor 2 (Nrf2)/Kelch Like ECH Associated Protein 1 (KEAP1) signaling pathway is the most important antioxidant stress pathway, we further analyzed the regulatory effect of SMP30 by WB to explore its molecular mechanism. Our study indicated that SMP30 may inhibit ROS accumulation, restore mitochondrial function, activate Nrf2/Keap1 signaling pathway, therefore protecting lens epithelial cells from oxidative stress-induced cell damage.


Subject(s)
Calcium-Binding Proteins , Cataract , Intracellular Signaling Peptides and Proteins , Mitochondria , Oxidative Stress , Apoptosis , Biomarkers/metabolism , Calcium-Binding Proteins/metabolism , Cataract/metabolism , Cataract/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Mitochondria/metabolism , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism
4.
Sci Rep ; 11(1): 13627, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34211074

ABSTRACT

Corneal innervation plays a major role in the pathobiology of diabetic corneal disease. However, innervation impact has mainly been investigated in the context of diabetic epitheliopathy and wound healing. Further studies are warranted in the corneal stroma-nerve interactions. This study unravels the nerve influence on corneal stroma metabolism. Corneal stromal cells were isolated from healthy (HCFs) and diabetes mellitus (Type1DM and Type2 DM) donors. Cells were cultured on polycarbonate membranes, stimulated by stable Vitamin C, and stroma-only and stroma-nerve co-cultures were investigated for metabolic alterations. Innervated compared to stroma-only constructs exhibited significant alterations in pyrimidine, glycerol phosphate shuttle, electron transport chain and glycolysis. The most highly altered metabolites between healthy and T1DMs innervated were phosphatidylethanolamine biosynthesis, and pyrimidine, methionine, aspartate metabolism. Healthy and T2DMs main pathways included aspartate, glycerol phosphate shuttle, electron transport chain, and gluconeogenesis. The metabolic impact on T1DMs and T2DMs was pyrimidine, purine, aspartate, and methionine. Interestingly, the glucose-6-phosphate and oxaloacetate was higher in T2DMs compared to T1DMs. Our in vitro co-culture model allows the examination of key metabolic pathways corresponding to corneal innervation in the diabetic stroma. These novel findings can pave the way for future studies to fully understand the metabolic distinctions in the diabetic cornea.


Subject(s)
Corneal Diseases/metabolism , Corneal Stroma/innervation , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Cell Line , Cells, Cultured , Corneal Diseases/etiology , Corneal Diseases/pathology , Corneal Stroma/metabolism , Corneal Stroma/pathology , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/pathology , Energy Metabolism , Glucose/metabolism , Humans , Metabolic Networks and Pathways , Metabolome , Nerve Tissue/metabolism , Nerve Tissue/pathology
5.
Exp Eye Res ; 208: 108617, 2021 07.
Article in English | MEDLINE | ID: mdl-34010603

ABSTRACT

Peroxisome Proliferator-Activated Receptors (PPARs) are a family of nuclear receptors that play essential roles in modulating cell differentiation, inflammation, and metabolism. Three subtypes of PPARs are known: PPAR-alpha (PPARα), PPAR-gamma (PPARγ), and PPAR-beta/delta (PPARß/δ). PPARα activation reduces lipid levels and regulates energy homeostasis, activation of PPARγ results in regulation of adipogenesis, and PPARß/δ activation increases fatty acid metabolism and lipolysis. PPARs are linked to various diseases, including but not limited to diabetes, non-alcoholic fatty liver disease, glaucoma and atherosclerosis. In the past decade, numerous studies have assessed the functional properties of PPARs in the eye and key PPAR mechanisms have been discovered, particularly regarding the retina and cornea. PPARγ and PPARα are well established in their functions in ocular homeostasis regarding neuroprotection, neovascularization, and inflammation, whereas PPARß/δ isoform function remains understudied. Naturally, studies on PPAR agonists and antagonists, associated with ocular pathology, have also gained traction with the development of PPAR synthetic ligands. Studies on PPARs has significantly influenced novel therapeutics for diabetic eye disease, ocular neuropathy, dry eye, and age-related macular degeneration (AMD). In this review, therapeutic potentials and implications will be highlighted, as well as reported adverse effects. Further investigations are necessary before any of the PPARs ligands can be utilized, in the clinics, to treat eye diseases. Future research on the prominent role of PPARs will help unravel the complex mechanisms involved in order to prevent and treat ocular diseases.


Subject(s)
Eye Diseases/metabolism , Lipid Metabolism/physiology , Peroxisome Proliferator-Activated Receptors/physiology , Animals , Homeostasis , Humans , Ligands
6.
Exp Eye Res ; 207: 108578, 2021 06.
Article in English | MEDLINE | ID: mdl-33864786

ABSTRACT

OBJECTIVE: To compare the biometric parameters provided by A-scan ultrasonography and the Lenstar optical biometer in guinea pig eyes, including anterior segment depth (ASD), lens thickness (LT), vitreous chamber depth (VCD), and axial length (AL), and differences of them between treated form deprivation (FD) eyes and untreated fellow eyes after 4 weeks of FD. METHODS: Three-week-old guinea pigs (N = 41) were subjected to biometric measurements before monocular FD (baseline) and after a 4-week FD. Statistical analyses including within-subject standard deviation (SDwithin), coefficient of variation (CV), and intraclass correlation coefficient (ICC), used to evaluate repeatability for both the A-scan ultrasonography and the Lenstar individually, and correlation and Bland-Altman analyses were used to assess agreement between the two methods. The absolute values of ASD, LT, VCD and AL as measured by the two devices were compared, and the differences of them between treated (T) and untreated fellow (F) eyes (ΔASD, ΔLT, ΔVCD and ΔAL) (Δ = T-F) were compared between the two devices after 4 weeks of FD. RESULTS: Measurements by the Lenstar (ICC: 0.923-0.994) were more repeatable than A-scan ultrasonography (ICC: 0.825-0.870). There was a high correlation for AL (r = 0.851, P < 0.001), a moderate correlation for VCD (r = 0.571, P < 0.001) and LT (r = 0.423, P < 0.001), and a low correlation for ASD (r = 0.230, P < 0.01) between the two devices. The values for ASD, VCD and AL measured by A-scan ultrasonography were larger than those measured by the Lenstar (all, P < 0.001), while LT provided by A-scan ultrasonography was much smaller than that of the Lenstar (P < 0.001). Bland-Altman plots showed poor agreement of absolute values of the four parameters between the two devices. Moreover, there was a high correlation between both methods for ΔAL (r = 0.704, P < 0.001), a moderate correlation for ΔVCD (r = 0.534, P < 0.001) and ΔASD (r = 0.574, P < 0.001), and no correlation for ΔLT (r = 0.303, P = 0.054). The ΔASD, ΔLT, and ΔAL measurements obtained by A-scan ultrasonography were greater than those obtained by the Lenstar (all, P < 0.001), while ΔVCD was mildly smaller using A-scan ultrasonography (P < 0.05). Bland-Altman plots illustrated there is good agreement of ΔAL, ΔVCD, ΔASD, and ΔLT between the two devices. CONCLUSIONS: The Lenstar exhibited better repeatability and provided smaller measurements for AL, VCD and ASD than A-scan ultrasonography. Furthermore, a high correlation and a good agreement for the ΔAL was observed between the two devices after a period of FD. In summary, the two devices cannot replace each other directly to obtain absolute values of ASD, LT, VCD and AL, but the Lenstar still can serve as an option in measuring ΔAL between eyes in guinea pig myopia model.


Subject(s)
Anterior Eye Segment/anatomy & histology , Axial Length, Eye/anatomy & histology , Biometry/instrumentation , Interferometry/methods , Lens, Crystalline/anatomy & histology , Ultrasonography/methods , Vitreous Body/anatomy & histology , Animals , Guinea Pigs , Light , Male , Reproducibility of Results
7.
J Cell Mol Med ; 25(6): 3063-3079, 2021 03.
Article in English | MEDLINE | ID: mdl-33599104

ABSTRACT

Primary open-angle glaucoma (POAG) is characterized by irreversible neurodegeneration accompanied by visual field defects and high intraocular pressure. Currently, an effective treatment is not available to prevent the progression of POAG, other than treatments to decrease the high intraocular pressure. We performed proteomic analysis of aqueous humour (AH) samples from patients with POAG combined with cataract and patients with cataract to obtain a better understanding of the pathogenesis of POAG and explore potential treatment targets for this condition. Samples were collected from 10 patients with POAG combined with cataract and 10 patients with cataract. Samples from each group were pooled. A high-resolution, label-free, liquid chromatography-tandem mass spectrometry-based quantitative proteomic analysis was performed. In total, 610 proteins were identified in human AH samples from the two groups. A total of 48 up-regulated proteins and 49 down-regulated proteins were identified in the POAG combined with cataract group compared with the control group. Gene Ontology (GO) analysis revealed key roles for these proteins in inflammation, immune responses, growth and development, cellular movement and vesicle-mediated transport in the biological process category. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis indicated the down-regulated expression of glutathione S-transferase P (GSTP1) in the glutathione metabolism signalling pathway in the POAG combined with cataract group. Additionally, certain significantly differentially expressed proteins in the proteomic profile were verified by enzyme-linked immunosorbent assay (ELISA). GSTP1 levels were reduced in the human AH samples from the POAG combined with cataract group, based on the results of ELISA and proteomic profiling. Therefore, GSTP1, a redox-related marker, may be involved in the pathological process of POAG and may become a treatment target in the future.


Subject(s)
Aqueous Humor/metabolism , Biomarkers , Gene Expression , Glaucoma, Open-Angle/genetics , Glaucoma, Open-Angle/metabolism , Glutathione S-Transferase pi/genetics , Glutathione S-Transferase pi/metabolism , Aged , Chromatography, High Pressure Liquid , Computational Biology/methods , Female , Glaucoma, Open-Angle/diagnosis , Humans , Male , Mass Spectrometry , Protein Interaction Maps , Proteome , Proteomics/methods , Reproducibility of Results
8.
Diabetes ; 69(6): 1279-1291, 2020 06.
Article in English | MEDLINE | ID: mdl-32213513

ABSTRACT

The purpose of this study was to investigate the protective role of peroxisome proliferator-activated receptor α (PPARα) against diabetic keratopathy and corneal neuropathy. Corneal samples were obtained from human donors with and without diabetes. Streptozotocin-induced diabetic rats and mice were orally treated with PPARα agonist fenofibrate. As shown by immunohistochemistry and Western blotting, PPARα was downregulated in the corneas of humans with diabetes and diabetic rats. Immunostaining of ß-III tubulin demonstrated that corneal nerve fiber metrics were decreased significantly in diabetic rats and mice, which were partially prevented by fenofibrate treatment. As evaluated using a Cochet-Bonnet aesthesiometer, corneal sensitivity was significantly decreased in diabetic mice, which was prevented by fenofibrate. PPARα -/- mice displayed progressive decreases in the corneal nerve fiber density. Consistently, corneal sensitivity was decreased in PPARα -/- mice relative to wild-type mice by 21 months of age. Diabetic mice showed increased incidence of spontaneous corneal epithelial lesion, which was prevented by fenofibrate while exacerbated by PPARα knockout. Western blot analysis revealed significantly altered neurotrophic factor levels in diabetic rat corneas, which were partially restored by fenofibrate treatment. These results indicate that PPARα protects the corneal nerve from degeneration induced by diabetes, and PPARα agonists have therapeutic potential in the treatment of diabetic keratopathy.


Subject(s)
Cornea/innervation , Diabetes Mellitus, Experimental/pathology , Nerve Degeneration/metabolism , PPAR alpha/metabolism , Animals , Down-Regulation , Fenofibrate/pharmacology , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Hypolipidemic Agents/pharmacology , Male , Nerve Degeneration/drug therapy , PPAR alpha/genetics , Rats , Rats, Sprague-Dawley
9.
Sci Rep ; 9(1): 13808, 2019 09 24.
Article in English | MEDLINE | ID: mdl-31551458

ABSTRACT

Keratoconus (KC) and chronic diabetes mellitus (DM) are both associated with significant defects in the human corneal structure. Studies have long suggested that DM is linked to KC, mainly via the crosslinking mechanism, but scientific evidences are lacking. The role of altered systemic metabolism is well-established in both DM and KC with studies suggesting localized altered cellular metabolism leading to the development of corneal pathologies. We have previously characterized the metabolic defects associated with both conditions using targeted metabolomics. To compare metabolic differences between KC and DM-derived corneal fibroblasts, we performed a respective study of two cohorts of the KC and DM populations using a retrospective analysis of targeted metabolomics data. The goal of this study was to identify the group of differentially regulated metabolites, in KC versus DM, so that we may unravel the link between the two devastating corneal pathologies.


Subject(s)
Diabetes Mellitus/pathology , Keratoconus/etiology , Cornea/metabolism , Cornea/pathology , Diabetes Complications/metabolism , Diabetes Complications/pathology , Diabetes Mellitus/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Keratoconus/metabolism , Male , Metabolomics/methods , Middle Aged , Retrospective Studies
10.
Exp Eye Res ; 186: 107709, 2019 09.
Article in English | MEDLINE | ID: mdl-31238077

ABSTRACT

Currently, over 10 million people worldwide are affected by corneal blindness. Corneal trauma and disease can cause irreversible distortions to the normal structure and physiology of the cornea often leading to corneal transplantation. However, donors are in short supply and risk of rejection is an ever-present concern. Although significant progress has been made in recent years, the wound healing cascade remains complex and not fully understood. Tissue engineering and regenerative medicine are currently at the apex of investigation in the pursuit of novel corneal therapeutics. This review uniquely integrates the clinical and cellular aspects of both corneal trauma and disease and provides a comprehensive view of the most recent findings and potential therapeutics aimed at restoring corneal homeostasis.


Subject(s)
Corneal Diseases , Corneal Injuries , Corneal Diseases/physiopathology , Corneal Diseases/therapy , Corneal Injuries/physiopathology , Corneal Injuries/therapy , Corneal Transplantation/methods , Humans , Ophthalmologic Surgical Procedures/trends , Stem Cell Transplantation/methods
SELECTION OF CITATIONS
SEARCH DETAIL