Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Antioxidants (Basel) ; 13(4)2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38671870

ABSTRACT

Dry eye disease (DED) is a multifactorial condition affecting the ocular surface. It is characterized by loss of tear film homeostasis and accompanied by ocular symptoms that may potentially result in damage to the ocular surface and even vision loss. Unmodifiable risk factors for DED mainly include aging, hormonal changes, and lifestyle issues such as reduced sleep duration, increased screen exposure, smoking, and ethanol consumption. As its prevalence continues to rise, DED has garnered considerable attention, prompting the exploration of potential new therapeutic targets. Recent studies have found that when the production of ROS exceeds the capacity of the antioxidant defense system on the ocular surface, oxidative stress ensues, leading to cellular apoptosis and further oxidative damage. These events can exacerbate inflammation and cellular stress responses, further increasing ROS levels and promoting a vicious cycle of oxidative stress in DED. Therefore, given the central role of reactive oxygen species in the vicious cycle of inflammation in DED, strategies involving antioxidants have emerged as a novel approach for its treatment. This review aims to enhance our understanding of the intricate relationship between oxidative stress and DED, thereby providing directions to explore innovative therapeutic approaches for this complex ocular disorder.

2.
Ocul Surf ; 32: 154-165, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38490475

ABSTRACT

Meibomian gland dysfunction (MGD) is a chronic abnormality of the Meibomian glands (MGs) that is recognized as the leading cause of evaporative dry eye worldwide. Despite its prevalence, however, the pathophysiology of MGD remains elusive, and effective disease management continues to be a challenge. In the past 50 years, different models have been developed to illustrate the pathophysiological nature of MGD and the underlying disease mechanisms. An understanding of these models is crucial if researchers are to select an appropriate model to address specific questions related to MGD and to develop new treatments. Here, we summarize the various models of MGD, discuss their applications and limitations, and provide perspectives for future studies in the field.


Subject(s)
Meibomian Gland Dysfunction , Meibomian Glands , Meibomian Gland Dysfunction/physiopathology , Meibomian Gland Dysfunction/metabolism , Meibomian Gland Dysfunction/therapy , Humans , Meibomian Glands/physiopathology , Meibomian Glands/metabolism , Animals , Tears/metabolism , Tears/physiology , Dry Eye Syndromes/physiopathology , Dry Eye Syndromes/metabolism , Disease Models, Animal
3.
Ocul Surf ; 29: 406-415, 2023 07.
Article in English | MEDLINE | ID: mdl-37327868

ABSTRACT

PURPOSE: To determine pathological changes of meibomian glands (MGs) after transient exposure of the rat eyelid margin to alkali solution. METHODS: Filter paper infiltrated with 1 N sodium hydroxide solution was applied to the eyelid margin of Sprague-Dawley rats for 30 s under general anesthesia, without touching the conjunctiva, after which the ocular surface and eyelid margin were examined by slit-lamp microscopy. In vivo confocal microscopy and stereomicroscopy were subsequently applied to observe MG morphology on day 5, day 10 and day 30 post alkali injury. Eyelid cross-sections were processed for H&E staining, Oil red O staining and immunofluorescent staining. RESULTS: After alkali injury, there was marked plugging of MG orifices, telangiectasia and hypertrophy of the eyelid margin, while corneal epithelium was intact at post-injury days 5 and 10. However, 30 days after alkali injury, mild corneal epithelial damage was observed. Degeneration of MG acini was observed at days 5 and became aggravated at days 10 and 30, along with MG duct dilation and acini loss. Oil red O staining showed lipid accumulation in the dilated duct. Inflammatory cell infiltration and the presence of apoptotic cells was seen in the MG loci 5 days post injury, but diminished at days 10 and 30. Cytokeratin 10 expression was increased in dilated duct, while cytokeratin 14, PPAR-γ, Ki67 and LRIG1 expression were decreased in the acini of injured loci. CONCLUSIONS: Transitory alkali exposure of the rat eyelid margin obstructs the MG orifice and induces pathological changes of MG dysfunction.


Subject(s)
Corneal Injuries , Eyelid Diseases , Meibomian Gland Dysfunction , Animals , Rats , Meibomian Glands/metabolism , Meibomian Gland Dysfunction/metabolism , Eyelid Diseases/metabolism , Rats, Sprague-Dawley , Corneal Injuries/metabolism , Alkalies/toxicity , Alkalies/metabolism , Tears/metabolism
4.
Invest Ophthalmol Vis Sci ; 63(1): 30, 2022 01 03.
Article in English | MEDLINE | ID: mdl-35072689

ABSTRACT

Purpose: Patients diagnosed with diabetes are inclined to have abnormalities on stability of tear film and disorder of meibomian gland (MG). This study aims to explore the pathological change of MG induced by diabetes in a rat model. Methods: Sprague-Dawley (SD) rats were intraperitoneally injected with streptozotocin (STZ) to establish a diabetic animal model. Lipid accumulation in MG was detected by Oil Red O staining and LipidTox staining. Cell proliferation status was determined by Ki67 and P63 immunostaining, whereas cell apoptosis was confirmed by TUNEL assay. Gene expression of inflammatory cytokines and adhesion molecules IL-1α, IL-1ß, ELAM1, ICAM1, and VCAM1 were detected by RT-PCR. Activation of ERK, NF-κB, and AMPK signaling pathways was determined by Western Blot analysis. Oxidative stress-related factors NOX4, 4HNE, Nrf2, HO-1, and SOD2 were detected by immunostaining or Western Blot analysis. Tom20 and Tim23 immunostaining and transmission electron microscopy were performed to evaluate the mitochondria functional and structure change. Results: Four months after STZ injection, there was acini dropout in MG of diabetic rats. Evident infiltration of inflammatory cells, increased expression of inflammatory factors, and adhesion molecules, as well as activated ERK and NF-κB signaling pathways were identified. Oxidative stress of MG was evident in 4-month diabetic rats. Phospho-AMPK was downregulated in MG of 2-month diabetic rats and more prominent in 4-month rats. After metformin treatment, phospho-AMPK was upregulated and the morphology of MG was well maintained. Moreover, inflammation and oxidative stress of MG were alleviated after metformin intervention. Conclusions: Long-term diabetes may lead to Meibomian gland dysfunction (MGD). AMPK may be a therapeutic target of MGD induced by diabetes.


Subject(s)
Blood Glucose/metabolism , Cytokines/metabolism , Hyperglycemia/complications , Meibomian Gland Dysfunction/etiology , Meibomian Glands/metabolism , Animals , Disease Models, Animal , Hyperglycemia/metabolism , Male , Meibomian Gland Dysfunction/metabolism , Oxidative Stress , Rats , Rats, Sprague-Dawley , Signal Transduction
5.
Invest Ophthalmol Vis Sci ; 62(10): 13, 2021 08 02.
Article in English | MEDLINE | ID: mdl-34398199

ABSTRACT

Purpose: To determine if a high-fat diet (HFD) induces meibomian gland (MG) inflammation in mice. Methods: Male C57BL/6J mice were fed a standard diet (SD), HFD, or HFD supplemented with the peroxisome proliferator-activated receptor gamma (PPAR-γ) agonist rosiglitazone for various durations. Body weight, blood lipid levels, and eyelid changes were monitored at regular intervals. MG sections were subjected to hematoxylin and eosin staining, LipidTox staining, TUNEL assay, and immunostaining. Quantitative RT-PCR and western blot analyses were performed to detect relative gene expression and signaling pathway activation in MGs. Results: MG acinus accumulated more lipids in the mice fed the HFD. Periglandular CD45-positive and F4/80-positive cell infiltration were more evident in the HFD mice, and they were accompanied by upregulation of inflammation-related cytokines. PPAR-γ downregulation accompanied activation of the mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB) signaling pathways in the HFD mice. There was increased acini cell apoptosis and mitochondria damage in mice fed the HFD. MG inflammation was ameliorated following a shift to the standard diet and rosiglitazone treatment in the mice fed the HFD. Conclusions: HFD-induced declines in PPAR-γ expression and MAPK and NF-κB signaling pathway activation resulted in MG inflammation and dysfunction in mice.


Subject(s)
Diet, High-Fat/adverse effects , Inflammation/metabolism , Meibomian Glands/pathology , Mitogen-Activated Protein Kinase Kinases/biosynthesis , NF-kappa B/biosynthesis , Uveitis/metabolism , Animals , Biomarkers/metabolism , Disease Models, Animal , Inflammation/pathology , Male , Meibomian Glands/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction , Uveitis/etiology , Uveitis/pathology
6.
Am J Pathol ; 190(12): 2387-2402, 2020 12.
Article in English | MEDLINE | ID: mdl-32919976

ABSTRACT

The lacrimal gland is critical for maintaining the homeostasis of the ocular surface microenvironment through secreting aqueous tears in mammals. Many systemic diseases such as Sjögren syndrome, rheumatoid arthritis, and diabetes can alter the lacrimal gland function, eventually resulting in aqueous tear-deficient dry eye. Here, a high-fat diet (HFD) experimental mouse model was used to clarify how hyperlipidemia affects lacrimal gland function. Aqueous tear secretion fell about 50% after 1 month on a HFD. Lipid droplets accumulated in the matrix and acinar cells of the lacrimal gland after this period, along with changes in the lipid metabolism, changes in gene expression levels, and disruption of fatty acid oxidative activity. Immune cell infiltration and rises in the gene expression levels of the inflammation-related cytokines Il1ß, Tnfα, Tsg6, Il10, Mmp2, and Mmp9 were found. HFD also induced mitochondrial hypermegasoma, increased apoptosis, and decreased lacrimal gland acinar cell proliferation. Replacement of the HFD with the standard diet partially reversed pathologic changes in the lacrimal gland. Similarly, supplementing the HFD with fenofibrate also partially reversed the inhibited tear secretion and reduced lipid accumulation, inflammation, and oxidative stress levels. The authors conclude that a HFD induces pathophysiological changes and functional decompensation of the lacrimal gland. Therefore, ingestion of a HFD may be a causative factor of dry eye disease.


Subject(s)
Diet, High-Fat , Dry Eye Syndromes/drug therapy , Lacrimal Apparatus/pathology , Sjogren's Syndrome/drug therapy , Animals , Cytokines/metabolism , Disease Models, Animal , Dry Eye Syndromes/metabolism , Lacrimal Apparatus/metabolism , Male , Mice, Inbred C57BL , Mitochondria/metabolism , Tears/drug effects , Tears/metabolism
7.
Ocul Surf ; 18(2): 267-276, 2020 04.
Article in English | MEDLINE | ID: mdl-32120007

ABSTRACT

PURPOSE: A high-fat diet leads to dysfunction in multiple systems of the body. Herein we investigate the effects of a high-fat diet on the ocular surface using a murine model. METHODS: Four-week-old male C57BL/6 mice were fed with a standard-fat diet (10 kcal% fat, SFD) or a high-fat diet (60 kcal% fat, HFD) for 1 or 3 months. Phenol red thread test was used to detect tear production, oregon green dextran (OGD) staining was performed to assess corneal epithelial permeability, and PAS staining was conducted to ascertain the presence of conjunctival goblet cells. Squamous metaplasia in the ocular surface and corneal epithelial barrier function were detected by immunofluorescent staining, zymography and Western blot analysis. Oxidative stress related protein expression was evaluated by immunostaining and Western blot analysis. Corneal and conjunctival cell apoptosis was determined by TUNEL assay and caspase-3 expression. RESULTS: A HFD induced obvious ocular surface damages, including decreased tear production, notable OGD staining and distinct goblet cell loss. It also resulted in corneal epithelial barrier dysfunction and significant squamous metaplasia of the corneal and conjunctival epithelia. The HFD also upregulated key factors that regulate oxidative stress in the ocular surface, and upregulated cell apoptosis in ocular surface epithelial cells. CONCLUSIONS: A HFD induces dry eye-like ocular surface damages in mice via the activation of oxidative stress and an induction of apoptosis in the cells of the ocular surface.


Subject(s)
Dry Eye Syndromes , Epithelium, Corneal , Animals , Conjunctiva , Diet, High-Fat/adverse effects , Disease Models, Animal , Dry Eye Syndromes/etiology , Male , Mice , Mice, Inbred C57BL , Tears
8.
Am J Pathol ; 190(3): 563-576, 2020 03.
Article in English | MEDLINE | ID: mdl-31945314

ABSTRACT

Hyperlipidemia impacts on various diseases, such as atherosclerosis, hypertension, and diabetes mellitus. However, its influence, if any, on ocular tissues is largely unknown. Herein, we developed hyperlipidemic murine models by feeding 4-week-old male wild-type mice with a high-fat diet and apolipoprotein E knockout (ApoE-/-) mice with a high-fat diet or standard diet to investigate the corneal endothelial change under hyperlipidemic conditions. Oil Red O staining showed an accumulation of lipid droplets in corneal endothelial cells (CECs) of hyperlipidemic mice. Other manifestations included a reduced cell density and distorted cell morphology, a disruption of the endothelial cell tight junctions and adhesion junctions, a reduced number of surface microvilli, down-regulation of Na+-K+-ATPase expression and function, activation of oxidative stress, changes in mitochondrial ultrastructure, and increased apoptosis. CEC recovery after injury, moreover, was diminished in hyperlipidemic mice; and high palmitate levels were found in the aqueous humor. In vitro hyperlipemia model, moreover, was found to be associated with dose-dependent CEC cytotoxicity, altered cell morphology, reduced pump function, and an induction of oxidative stress, leading to functional and pathologic changes in the corneal endothelium.


Subject(s)
Apolipoproteins E/genetics , Diet, High-Fat/adverse effects , Hyperlipidemias/complications , Oxidative Stress , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Apoptosis , Cell Survival , Disease Models, Animal , Dose-Response Relationship, Drug , Down-Regulation , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Corneal/metabolism , Endothelium, Corneal/pathology , Hyperlipidemias/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Mitochondria/ultrastructure , Palmitates/toxicity , Sodium-Potassium-Exchanging ATPase/genetics , Tight Junctions/metabolism , Tight Junctions/pathology
9.
Ocul Surf ; 17(4): 777-786, 2019 10.
Article in English | MEDLINE | ID: mdl-31201956

ABSTRACT

PURPOSE: To investigate the pathological changes of the meibomian gland (MG) and ocular surface in Apolipoprotein E knockout (ApoE-/-) mice and to investigate the association of meibomian gland dysfunction (MGD) with hyperlipidemia. METHODS: Total plasma cholesterol was measured in different ages of ApoE-/- and wild type (WT) mice, whilst the ocular surfaces were observed by slit-lamp biomicroscopy. MG sections were subjected to H&E staining, Oil Red O staining, TUNEL assay and immunostaining. Quantitate RT-PCR and Western blot analyses were performed to detect the relative gene expression in MGs. The 5-month-old ApoE-/- mice were administered with rosiglitazone or GW9662 + rosiglitazone via oral gavage for 2 months to determine their effect on MG pathological change. RESULTS: We found eyelid abnormality, MG dropout, abnormal MG acinar morphology, dilated MG duct and plugging of the MG orifice in ApoE-/- mice. MG acini in ApoE-/- mice showed exaggerated lipid accumulation. Abnormal keratinization increased in MG duct, accompanied with decreased proliferation and increased apoptosis in ApoE-/- mice. Inflammatory cells infiltrated into the surrounding microenvironment of MG acini, and the NF-κB signaling pathway was activated in MG acinar cells. Oxidative stress was evident in MG acinar cells of ApoE-/- mice. Further investigation showed downregulation of PPAR-γ in MG acinar cells of ApoE-/- mice. PPAR-γ agonist rosiglitazone treatment reduced the morbidity of eyelid, as well as corneal pathological changes and MG inflammation in ApoE-/- mice. CONCLUSION: MGD and hyperlipidemia are closely associated in ApoE-/- mice, which represent a new model to study the pathophysiology of MGD related to dyslipidemia.


Subject(s)
Apolipoproteins E/genetics , Gene Expression Regulation , Hyperlipidemias/complications , Meibomian Gland Dysfunction/etiology , Meibomian Glands/pathology , RNA/genetics , Animals , Apolipoproteins E/biosynthesis , Apolipoproteins E/blood , Blotting, Western , Disease Models, Animal , Hyperlipidemias/metabolism , Male , Meibomian Gland Dysfunction/diagnosis , Meibomian Gland Dysfunction/metabolism , Meibomian Glands/metabolism , Mice , Mice, Knockout , Rabbits
10.
Invest Ophthalmol Vis Sci ; 58(11): 4898-4907, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28973340

ABSTRACT

Purpose: To investigate the role of mitogen-activated protein kinase-activated protein kinase-2 (MK2) in ocular surface damage of dry eye. Methods: MK2 inhibition was performed in mice subjected to desiccating stress (DS) by topical application of MK2 inhibitor (MK2i) or vehicle eye drops. The total and phosphorylated MK2 in conjunctiva were detected by Western blot. The phenol red cotton test was used to measure tear production, and Oregon green dextran staining was performed to assess corneal epithelial barrier function. PAS staining was used to quantify conjunctival goblet cells. Immunofluorescent staining and quantitative RT-PCR were used to assess the expression of matrix metalloproteinase (MMP)-3 and -9 in corneal epithelium. Apoptosis in ocular surface was assessed by TUNEL and immunofluorescent staining for activated caspase-3 and -8. Inflammation was evaluated by CD4+ T-cell infiltration and production of T helper (Th) cytokines, including IFN-γ, IL-13, and IL-17A in conjunctiva. Results: DS promoted MK2 activation in conjunctiva. Compared with vehicle control mice, MK2i-treated mice showed increased tear production, decreased goblet cell loss, and improved corneal barrier function. Topical MK2 inhibition decreased the expression of MMP-3 and -9 in corneal epithelium, and suppressed cell apoptosis in ocular surface under DS. Topical MK2 inhibition decreased CD4+ T-cell infiltration, with decreased production of IFN-γ and IL-17A and increased production of IL-13 in conjunctiva. Conclusions: Topical MK2 inhibition effectively alleviated ocular surface damage via suppressing cell apoptosis and CD4+ T-cell-mediated inflammation in ocular surface of dry eye.


Subject(s)
Conjunctiva/drug effects , Dry Eye Syndromes/drug therapy , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/physiology , Peptides/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/physiology , Animals , Apoptosis/drug effects , Blotting, Western , Conjunctiva/metabolism , Cytokines/metabolism , Disease Models, Animal , Dry Eye Syndromes/metabolism , Epithelium, Corneal/drug effects , Epithelium, Corneal/metabolism , Goblet Cells/metabolism , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Tears/metabolism
11.
Exp Eye Res ; 163: 29-36, 2017 10.
Article in English | MEDLINE | ID: mdl-28950937

ABSTRACT

The meibomian gland is the major contributor to the tear film lipid layer. It is generally accepted that meibomian gland secretions, i.e, meibum, play a critical role in the homeostasis of the tear film. Lipid components of meibum and their structure, as well as functions were intensively studied. However, the proteins from meibum have not attracted enough attention. This review summarizes current knowledge about protein components of the meibum, particularly their function on tear film and ocular surface, and changes in the proteins during meibomian gland dysfunction (MGD).


Subject(s)
Eye Proteins/physiology , Meibomian Glands/physiology , Tears/chemistry , Eye Proteins/metabolism , Eyelid Diseases/metabolism , Eyelid Diseases/physiopathology , Humans , Meibomian Glands/metabolism , Meibomian Glands/physiopathology
12.
J Biol Chem ; 292(32): 13391-13401, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28655773

ABSTRACT

The EDA gene encodes ectodysplasin A (Eda), which if mutated causes X-linked hypohidrotic ectodermal dysplasia (XLHED) disease in humans. Ocular surface changes occur in XLHED patients whereas its underlying mechanism remains elusive. In this study, we found Eda was highly expressed in meibomian glands, and it was detected in human tears but not serum. Corneal epithelial integrity was defective and the thickness was reduced in the early postnatal stage of Eda mutant Tabby mice. Corneal epithelial cell proliferation decreased and the epithelial wound healing was delayed in Tabby mice, whereas it was restored by exogenous Eda. Eda exposure promoted mouse corneal epithelial wound healing during organ culture, whereas scratch wound assay showed that it did not affect human corneal epithelial cell line migration. Epidermal growth factor receptor (EGFR), phosphorylated EGFR (p-EGFR), and phosphorylated ERK1/2 (p-ERK) were down-regulated in Tabby mice corneal epithelium. Eda treatment up-regulated the expression of Ki67, EGFR, p-EGFR, and p-ERK in human corneal epithelial cells in a dose-dependent manner. In conclusion, Eda protein can be secreted from meibomian glands and promotes corneal epithelial cell proliferation through regulation of the EGFR signaling pathway. Eda release into the tears plays an essential role in the maintenance of corneal epithelial homeostasis.


Subject(s)
Ectodermal Dysplasia 1, Anhidrotic/metabolism , Ectodysplasins/metabolism , Epithelium, Corneal/metabolism , Eyelid Diseases/metabolism , Meibomian Glands/metabolism , Adolescent , Adult , Animals , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Ectodermal Dysplasia 1, Anhidrotic/drug therapy , Ectodermal Dysplasia 1, Anhidrotic/pathology , Ectodermal Dysplasia 1, Anhidrotic/physiopathology , Ectodysplasins/genetics , Ectodysplasins/pharmacology , Ectodysplasins/therapeutic use , Epithelium, Corneal/drug effects , Epithelium, Corneal/injuries , Epithelium, Corneal/pathology , ErbB Receptors/metabolism , Eyelid Diseases/pathology , Eyelid Diseases/physiopathology , Female , Humans , Male , Meibomian Glands/pathology , Meibomian Glands/physiopathology , Mice, Mutant Strains , Organ Culture Techniques , Phosphorylation , Protein Processing, Post-Translational , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Signal Transduction , Tears/metabolism , Wound Healing/drug effects , Young Adult
13.
Int J Mol Sci ; 18(7)2017 Jun 29.
Article in English | MEDLINE | ID: mdl-28661456

ABSTRACT

Dry eye can damage the ocular surface and result in mild corneal epithelial defect to blinding corneal pannus formation and squamous metaplasia. Significant progress in the treatment of dry eye has been made in the last two decades; progressing from lubricating and hydrating the ocular surface with artificial tear to stimulating tear secretion; anti-inflammation and immune regulation. With the increase in knowledge regarding the pathophysiology of dry eye, we propose in this review the concept of ocular surface microenvironment. Various components of the microenvironment contribute to the homeostasis of ocular surface. Compromise in one or more components can result in homeostasis disruption of ocular surface leading to dry eye disease. Complete evaluation of the microenvironment component changes in dry eye patients will not only lead to appropriate diagnosis, but also guide in timely and effective clinical management. Successful treatment of dry eye should be aimed to restore the homeostasis of the ocular surface microenvironment.


Subject(s)
Cellular Microenvironment , Dry Eye Syndromes/pathology , Dry Eye Syndromes/therapy , Eye/pathology , Cellular Microenvironment/drug effects , Conjunctiva/drug effects , Conjunctiva/pathology , Cornea/drug effects , Cornea/pathology , Dry Eye Syndromes/drug therapy , Eye/drug effects , Eye/immunology , Eye/microbiology , Eyelids/drug effects , Eyelids/pathology , Homeostasis , Hormones , Humans , Lacrimal Apparatus/drug effects , Lacrimal Apparatus/pathology , Lubricant Eye Drops/therapeutic use , Meibomian Glands/drug effects , Meibomian Glands/pathology , Microbiota , Tears/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...