Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 888
Filter
1.
Int J Mol Sci ; 25(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38731911

ABSTRACT

In drug discovery, selecting targeted molecules is crucial as the target could directly affect drug efficacy and the treatment outcomes. As a member of the CCN family, CTGF (also known as CCN2) is an essential regulator in the progression of various diseases, including fibrosis, cancer, neurological disorders, and eye diseases. Understanding the regulatory mechanisms of CTGF in different diseases may contribute to the discovery of novel drug candidates. Summarizing the CTGF-targeting and -inhibitory drugs is also beneficial for the analysis of the efficacy, applications, and limitations of these drugs in different disease models. Therefore, we reviewed the CTGF structure, the regulatory mechanisms in various diseases, and drug development in order to provide more references for future drug discovery.


Subject(s)
Connective Tissue Growth Factor , Drug Discovery , Humans , Connective Tissue Growth Factor/metabolism , Drug Discovery/methods , Animals , Neoplasms/drug therapy , Neoplasms/metabolism , Eye Diseases/drug therapy , Eye Diseases/metabolism , Fibrosis , Nervous System Diseases/drug therapy , Nervous System Diseases/metabolism , Gene Expression Regulation/drug effects
2.
J Histochem Cytochem ; 72(5): 329-352, 2024 05.
Article in English | MEDLINE | ID: mdl-38733294

ABSTRACT

Lactate and ATP formation by aerobic glycolysis, the Warburg effect, is considered a hallmark of cancer. During angiogenesis in non-cancerous tissue, proliferating stalk endothelial cells (ECs) also produce lactate and ATP by aerobic glycolysis. In fact, all proliferating cells, both non-cancer and cancer cells, need lactate for the biosynthesis of building blocks for cell growth and tissue expansion. Moreover, both non-proliferating cancer stem cells in tumors and leader tip ECs during angiogenesis rely on glycolysis for pyruvate production, which is used for ATP synthesis in mitochondria through oxidative phosphorylation (OXPHOS). Therefore, aerobic glycolysis is not a specific hallmark of cancer but rather a hallmark of proliferating cells and limits its utility in cancer therapy. However, local treatment of angiogenic eye conditions with inhibitors of glycolysis may be a safe therapeutic option that warrants experimental investigation. Most types of cells in the eye such as photoreceptors and pericytes use OXPHOS for ATP production, whereas proliferating angiogenic stalk ECs rely on glycolysis for lactate and ATP production. (J Histochem Cytochem XX.XXX-XXX, XXXX).


Subject(s)
Adenosine Triphosphate , Neoplasms , Neovascularization, Pathologic , Humans , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/biosynthesis , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/blood supply , Neoplasms/drug therapy , Animals , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Glycolysis , Eye Diseases/metabolism , Eye Diseases/pathology , Oxidative Phosphorylation
3.
Biochem Biophys Res Commun ; 717: 150041, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38710142

ABSTRACT

Ocular inflammation-associated diseases are leading causes of global visual impairment, with limited treatment options. Adiponectin, a hormone primarily secreted by adipose tissue, binds to its receptors, which are widely distributed throughout the body, exerting powerful physiological regulatory effects. The protective role of adiponectin in various inflammatory diseases has gained increasing attention in recent years. Previous studies have confirmed the presence of adiponectin and its receptors in the eyes. Furthermore, adiponectin and its analogs have shown potential as novel drugs for the treatment of inflammatory eye diseases. This article summarizes the evidence for the interplay between adiponectin and inflammatory eye diseases and provides new perspectives on the diagnostic and therapeutic possibilities of adiponectin.


Subject(s)
Adiponectin , Inflammation , Receptors, Adiponectin , Signal Transduction , Humans , Adiponectin/metabolism , Receptors, Adiponectin/metabolism , Animals , Inflammation/metabolism , Eye Diseases/metabolism , Eye Diseases/drug therapy
4.
Adv Clin Chem ; 120: 69-115, 2024.
Article in English | MEDLINE | ID: mdl-38762243

ABSTRACT

An extensive exploration of lacrimal fluid molecular biomarkers in understanding and diagnosing a spectrum of ocular and systemic diseases is presented. The chapter provides an overview of lacrimal fluid composition, elucidating the roles of proteins, lipids, metabolites, and nucleic acids within the tear film. Pooled versus single-tear analysis is discussed to underline the benefits and challenges associated with both approaches, offering insights into optimal strategies for tear sample analysis. Subsequently, an in-depth analysis of tear collection methods is presented, with a focus on Schirmer's test strips and microcapillary tubes methods. Alternative tear collection techniques are also explored, shedding light on their applicability and advantages. Variability factors, including age, sex, and diurnal fluctuations, are examined in the context of their impact on tear biomarker analysis. The main body of the chapter is dedicated to discussing specific biomarkers associated with ocular discomfort and a wide array of ocular diseases. From dry eye disease and thyroid-associated ophthalmopathy to keratoconus, age-related macular degeneration, diabetic retinopathy, and glaucoma, the intricate relationship between molecular biomarkers and these conditions is thoroughly dissected. Expanding beyond ocular pathologies, the chapter explores the applicability of tear biomarkers in diagnosing systemic diseases such as multiple sclerosis, amyotrophic lateral sclerosis, Alzheimer's disease, Parkinson's disease, and cancer. This broader perspective underscores the potential of lacrimal fluid analysis in offering non-invasive diagnostic tools for conditions with far-reaching implications.


Subject(s)
Biomarkers , Tears , Humans , Tears/metabolism , Tears/chemistry , Biomarkers/analysis , Biomarkers/metabolism , Eye Diseases/diagnosis , Eye Diseases/metabolism
5.
Ageing Res Rev ; 97: 102308, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38615894

ABSTRACT

Aging entails the deterioration of the body's organs, including overall damages at both the genetic and cellular levels. The prevalence of age-related ocular disease such as macular degeneration, dry eye diseases, glaucoma and cataracts is increasing as the world's population ages, imposing a considerable economic burden on individuals and society. The development of age-related ocular disease is predominantly triggered by oxidative stress and chronic inflammatory reaction. Heme oxygenase-1 (HO-1) is a crucial antioxidant that mediates the degradative process of endogenous iron protoporphyrin heme. It catalyzes the rate-limiting step of the heme degradation reaction, and releases the metabolites such as carbon monoxide (CO), ferrous, and biliverdin (BV). The potent scavenging activity of these metabolites can help to defend against peroxides, peroxynitrite, hydroxyl, and superoxide radicals. Other than directly decomposing endogenous oxidizing substances (hemoglobin), HO-1 is also a critical regulator of inflammatory cells and tissue damage, exerting its anti-inflammation activity through regulating complex inflammatory networks. Therefore, promoting HO-1 expression may act as a promising therapeutic strategy for the age-related ocular disease. However, emerging evidences suggest that the overexpression of HO-1 significantly contributes to ferroptosis due to its dual nature. Surplus HO-1 leads to excessive Fe2+ and reactive oxygen species, thereby causing lipid peroxidation and ferroptosis. In this review, we elucidate the role of HO-1 in countering age-related disease, and summarize recent pharmacological trials that targeting HO-1 for disease management. Further refinements of the knowledge would position HO-1 as a novel therapeutic target for age-related ocular disease.


Subject(s)
Aging , Eye Diseases , Heme Oxygenase-1 , Humans , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Aging/metabolism , Aging/genetics , Eye Diseases/metabolism , Animals , Oxidative Stress/physiology
6.
Prog Retin Eye Res ; 100: 101249, 2024 May.
Article in English | MEDLINE | ID: mdl-38430990

ABSTRACT

Translocator protein (18 kDa) (Tspo), formerly known as peripheral benzodiazepine receptor is a highly conserved transmembrane protein primarily located in the outer mitochondrial membrane. In the central nervous system (CNS), especially in glia cells, Tspo is upregulated upon inflammation. Consequently, Tspo was used as a tool for diagnostic in vivo imaging of neuroinflammation in the brain and as a potential therapeutic target. Several synthetic Tspo ligands have been explored as immunomodulatory and neuroprotective therapy approaches. Although the function of Tspo and how its ligands exert these beneficial effects is not fully clear, it became a research topic of interest, especially in ocular diseases in the past few years. This review summarizes state-of-the-art knowledge of Tspo expression and its proposed functions in different cells of the retina including microglia, retinal pigment epithelium and Müller cells. Tspo is involved in cytokine signaling, oxidative stress and reactive oxygen species production, calcium signaling, neurosteroid synthesis, energy metabolism, and cholesterol efflux. We also highlight recent developments in preclinical models targeting Tspo and summarize the relevance of Tspo biology for ocular and retinal diseases. We conclude that glial upregulation of Tspo in different ocular pathologies and the use of Tspo ligands as promising therapeutic approaches in preclinical studies underline the importance of Tspo as a potential disease-modifying protein.


Subject(s)
Receptors, GABA , Retina , Humans , Receptors, GABA/metabolism , Animals , Retina/metabolism , Eye Diseases/metabolism , Retinal Diseases/metabolism , Microglia/metabolism
7.
Nat Commun ; 15(1): 1600, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38383453

ABSTRACT

Cross-species genome comparisons have revealed a substantial number of ultraconserved non-coding elements (UCNEs). Several of these elements have proved to be essential tissue- and cell type-specific cis-regulators of developmental gene expression. Here, we characterize a set of UCNEs as candidate CREs (cCREs) during retinal development and evaluate the contribution of their genomic variation to rare eye diseases, for which pathogenic non-coding variants are emerging. Integration of bulk and single-cell retinal multi-omics data reveals 594 genes under potential cis-regulatory control of UCNEs, of which 45 are implicated in rare eye disease. Mining of candidate cis-regulatory UCNEs in WGS data derived from the rare eye disease cohort of Genomics England reveals 178 ultrarare variants within 84 UCNEs associated with 29 disease genes. Overall, we provide a comprehensive annotation of ultraconserved non-coding regions acting as cCREs during retinal development which can be targets of non-coding variation underlying rare eye diseases.


Subject(s)
Eye Diseases , Multiomics , Humans , Retina/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Genome , Eye Diseases/genetics , Eye Diseases/metabolism
8.
Vet Ophthalmol ; 27(2): 104-113, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37246963

ABSTRACT

BACKGROUND: The ocular side effects of cancer chemotherapeutic drugs are relatively uncommon. Nonetheless, the ocular system has a potentially high sensitivity to toxic substances. This study proposed a framework to assess the effect of vincristine chemotherapy on intraocular pressure, tear protein, and oxidative stress in canines with transmissible venereal tumor (TVT). METHODS: The study group comprised 10 dogs with TVT, whose diagnosis was based on cytology, and all dogs were treated with vincristine for 4 weeks. Each animal was given a complete ophthalmic examination, followed by a standard Schirmer tear test. Before and 20 min after administering vincristine, intraocular pressure (IOP) was measured in the eyes with a noncontact tonometer. At any of the times mentioned, tear samples were collected using the Schirmer test procedure and were subjected to protein analysis-oxidative stress index (OSI), total antioxidant capacity (TAC), total oxidant status (TOS), nitric oxide (NO), and malondialdehyde (MDA) were determined, and standard statistical analysis was applied. RESULTS: No significant differences were found in protein in tears, but mean Pre and Postinjection IOP revealed a significant decrease in the eyes each week. Also, results indicated significant differences in oxidative stress markers: increased OSI, NO, and MDA, and reduced TAC. CONCLUSION: The importance of an increase in oxidative stress levels in the tears of vincristine-treated patients should be taken seriously, as it appears to play a role in the pathogenesis of eye disease. Therefore, during the treatment weeks prior to prescribing vincristine, eye diseases should be evaluated and considered.


Subject(s)
Eye Diseases , Venereal Tumors, Veterinary , Humans , Animals , Dogs , Vincristine/adverse effects , Intraocular Pressure , Venereal Tumors, Veterinary/drug therapy , Venereal Tumors, Veterinary/metabolism , Venereal Tumors, Veterinary/pathology , Eye Diseases/metabolism , Eye Diseases/veterinary , Tears/metabolism , Oxidative Stress
9.
J Control Release ; 365: 448-468, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38013069

ABSTRACT

Nanoscale extracellular vesicles (EVs), consisting of exomers, exosomes and microvesicles/ectosomes, have been extensively investigated in the last 20 years, although their biological role is still something of a mystery. EVs are involved in the transfer of lipids, nucleic acids and proteins from donor to recipient cells or distant organs as well as regulating cell-cell communication and signaling. Thus, EVs are important in intercellular communication and this is not limited to sister cells, but may also mediate the crosstalk between different cell types even over long distances. EVs play crucial functions in both cellular homeostasis and the pathogenesis of diseases, and since their contents reflect the status of the donor cell, they represent an additional valuable source of information for characterizing complex biological processes. Recent advances in isolation and analytical methods have led to substantial improvements in both characterizing and engineering EVs, leading to their use either as novel biomarkers for disease diagnosis/prognosis or even as novel therapies. Due to their capacity to carry biomolecules, various EV-based therapeutic applications have been devised for several pathological conditions, including eye diseases. In the eye, EVs have been detected in the retina, aqueous humor, vitreous body and also in tears. Experiences with other forms of intraocular drug applications have opened new ways to use EVs in the treatment of retinal diseases. We here provide a comprehensive summary of the main in vitro, in vivo, and ex vivo literature-based studies on EVs' role in ocular physiological and pathological conditions. We have focused on age-related macular degeneration, diabetic retinopathy, glaucoma, which are common eye diseases leading to permanent blindness, if not treated properly. In addition, the putative use of EVs in retinitis pigmentosa and other retinopathies is discussed. Finally, we have reviewed the potential of EVs as therapeutic tools and/or biomarkers in the above-mentioned retinal disorders. Evidence emerging from experimental disease models and human material strongly suggests future diagnostic and/or therapeutic exploitation of these biological agents in various ocular disorders with a good possibility to improve the patient's quality of life.


Subject(s)
Extracellular Vesicles , Eye Diseases , Retinal Diseases , Humans , Quality of Life , Extracellular Vesicles/metabolism , Biomarkers/metabolism , Retina/metabolism , Retinal Diseases/drug therapy , Retinal Diseases/metabolism , Eye Diseases/drug therapy , Eye Diseases/metabolism
10.
Sci Rep ; 13(1): 22839, 2023 12 21.
Article in English | MEDLINE | ID: mdl-38129447

ABSTRACT

Goblet cells (GCs) in the conjunctiva are specialized epithelial cells secreting mucins for the mucus layer of protective tear film and playing immune tolerance functions for ocular surface health. Because GC loss is observed in various ocular surface diseases, GC examination is important for precision diagnosis. Moxifloxacin-based fluorescence microscopy (MBFM) was recently developed for non-invasive high-contrast GC visualization. MBFM showed promise for GC examination by high-speed large-area imaging and a robust analysis method is needed to provide GC information. In this study, we developed a deep learning framework for GC image analysis, named dual-channel attention U-Net (DCAU-Net). Dual-channel convolution was used both to extract the overall image texture and to acquire the GC morphological characteristics. A global channel attention module was adopted by combining attention algorithms and channel-wise pooling. DCAU-Net showed 93.1% GC segmentation accuracy and 94.3% GC density estimation accuracy. Further application to both normal and ocular surface damage rabbit models revealed the spatial variations of both GC density and size in normal rabbits and the decreases of both GC density and size in damage rabbit models during recovery after acute damage. The GC analysis results were consistent with histology. Together with the non-invasive high-contrast imaging method, DCAU-Net would provide GC information for the diagnosis of ocular surface diseases.


Subject(s)
Deep Learning , Eye Diseases , Lagomorpha , Animals , Rabbits , Goblet Cells/metabolism , Conjunctiva/pathology , Tears/metabolism , Eye Diseases/metabolism , Cell Count
11.
Adv Sci (Weinh) ; 10(30): e2302909, 2023 10.
Article in English | MEDLINE | ID: mdl-37653617

ABSTRACT

Retinal ischemia is involved in the occurrence and development of various eye diseases, including glaucoma, diabetic retinopathy, and central retinal artery occlusion. To the best of our knowledge, few studies have reported self-assembling peptide natural products for the suppression of ocular inflammation and oxidative stress. Herein, a self-assembling peptide GFFYE is designed and synthesized, which can transform the non-hydrophilicity of rhein into an amphiphilic sustained-release therapeutic agent, and rhein-based therapeutic nanofibers (abbreviated as Rh-GFFYE) are constructed for the treatment of retinal ischemia-reperfusion (RIR) injury. Rh-GFFYE significantly ameliorates oxidative stress and inflammation in an in vitro oxygen-glucose deprivation (OGD) model of retinal ischemia and a rat model of RIR injury. Rh-GFFYE also significantly enhances retinal electrophysiological recovery and exhibits good biocompatibility. Importantly, Rh-GFFYE also promotes the transition of M1-type macrophages to the M2 type, ultimately altering the pro-inflammatory microenvironment. Further investigation of the treatment mechanism indicates that Rh-GFFYE activates the PI3K/AKT/mTOR signaling pathway to reduce oxidative stress and inhibits the NF-κB and STAT3 signaling pathways to affect inflammation and macrophage polarization. In conclusion, the rhein-loaded nanoplatform alleviates RIR injury by modulating the retinal microenvironment. The findings are expected to promote the clinical application of hydrophobic natural products in RIR injury-associated eye diseases.


Subject(s)
Biological Products , Eye Diseases , Nanofibers , Reperfusion Injury , Rats , Animals , Microglia/metabolism , Nanofibers/therapeutic use , Phosphatidylinositol 3-Kinases , Oxidative Stress , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Macrophages/metabolism , Inflammation/metabolism , Eye Diseases/metabolism , Biological Products/metabolism , Peptides/metabolism , Ischemia
12.
Adv Drug Deliv Rev ; 200: 115005, 2023 09.
Article in English | MEDLINE | ID: mdl-37419213

ABSTRACT

Vision impairment and loss due to posterior segment ocular disorders, including age-related macular degeneration and diabetic retinopathy, are a rapidly growing cause of disability globally. Current treatments consist primarily of intravitreal injections aimed at preventing disease progression and characterized by high cost and repeated clinic visits. Nanotechnology provides a promising platform for drug delivery to the eye, with potential to overcome anatomical and physiological barriers to provide safe, effective, and sustained treatment modalities. However, there are few nanomedicines approved for posterior segment disorders, and fewer that target specific cells or that are compatible with systemic administration. Targeting cell types that mediate these disorders via systemic administration may unlock transformative opportunities for nanomedicine and significantly improve patient access, acceptability, and outcomes. We highlight the development of hydroxyl polyamidoamine dendrimer-based therapeutics that demonstrate ligand-free cell targeting via systemic administration and are under clinical investigation for treatment of wet age-related macular degeneration.


Subject(s)
Dendrimers , Eye Diseases , Macular Degeneration , Humans , Dendrimers/metabolism , Eye/metabolism , Drug Delivery Systems , Eye Diseases/metabolism , Macular Degeneration/drug therapy , Macular Degeneration/metabolism
13.
Ocul Surf ; 29: 388-397, 2023 07.
Article in English | MEDLINE | ID: mdl-37327869

ABSTRACT

Lumican is a keratan sulfate proteoglycan that belongs to the small leucine-rich proteoglycan family. Research has lifted the veil on the versatile roles of lumican in the pathogenesis of eye diseases. Lumican has pivotal roles in the maintenance of physiological tissue homogenesis and is often upregulated in pathological conditions, e.g., fibrosis, scar tissue formation in injured tissues, persistent inflammatory responses and immune anomaly, etc. Herein, we will review literature regarding the role of lumican in pathogenesis of inherited congenital and acquired eye diseases, e.g., cornea dystrophy, cataract, glaucoma and chorioretinal diseases, etc.


Subject(s)
Eye Diseases , Lumican , Humans , Chondroitin Sulfate Proteoglycans/physiology , Cornea/pathology , Eye Diseases/metabolism , Eye Diseases/pathology , Keratan Sulfate/physiology , Proteoglycans/physiology
14.
Exp Eye Res ; 233: 109557, 2023 08.
Article in English | MEDLINE | ID: mdl-37380095

ABSTRACT

As a water channel protein, aquaporin 5 (AQP5) is essential for the maintenance of the normal physiological functions of ocular tissues. This review provides an overview of the expression and function of AQP5 in the eye and discusses their role in related eye diseases. Although AQP5 plays a vital role in ocular functions, such as maintaining corneal and lens transparency, regulating water movement, and maintaining homeostasis, some of its functions in ocular tissues are still unclear. Based on the key role of AQP5 in eye function, this review suggests that in the future, eye diseases may be treated by regulating the expression of aquaporin.


Subject(s)
Eye Diseases , Lens, Crystalline , Humans , Aquaporin 5 , Cornea/metabolism , Lens, Crystalline/metabolism , Eye Diseases/metabolism , Ocular Physiological Phenomena
15.
Biomater Sci ; 11(13): 4490-4507, 2023 Jun 27.
Article in English | MEDLINE | ID: mdl-37222479

ABSTRACT

Numerous novel nano-based ocular drug delivery systems have been developed to overcome the limitations of conventional drug delivery systems, which have demonstrated promising results in ocular disease models and clinical practice. Of all the nano-based drug delivery systems approved or under clinical investigation, topical instillation of eye drops is the most common route for administering therapeutics to the eye. Although this pathway is a viable way of ocular drug delivery to treat many ocular diseases because of its potential to eliminate the risks of intravitreal injection and the toxicity of systemic drug delivery, it remains a major challenge to efficiently treat posterior ocular diseases through topical administration of eye drops. So far, relentless efforts have been dedicated to the development of novel nano-based drug delivery systems with the aim of possible clinical translation. They are designed or modified to facilitate drug delivery to the retina by increasing the retention time, promoting drug penetration across barriers, and targeting specific cells or tissues. In this paper, we provided a snapshot of nano-based drug delivery systems that are currently marketed and under investigation in clinical trials for the treatment of ocular diseases and highlighted some examples of recent preclinical research on novel nano-based systems as eye drops to the posterior segment of the eye.


Subject(s)
Eye Diseases , Humans , Eye Diseases/drug therapy , Eye Diseases/metabolism , Nanoparticle Drug Delivery System , Drug Delivery Systems/methods , Retina , Ophthalmic Solutions/therapeutic use
16.
Front Immunol ; 14: 1097456, 2023.
Article in English | MEDLINE | ID: mdl-36969177

ABSTRACT

Extracellular vesicles include exosomes, microvesicles, and apoptotic bodies. Their cargos contain a diverse variety of lipids, proteins, and nucleic acids that are involved in both normal physiology and pathology of the ocular system. Thus, studying extracellular vesicles may lead to a more comprehensive understanding of the pathogenesis, diagnosis, and even potential treatments for various diseases. The roles of extracellular vesicles in inflammatory eye disorders have been widely investigated in recent years. The term "inflammatory eye diseases" refers to a variety of eye conditions such as inflammation-related diseases, degenerative conditions with remarkable inflammatory components, neuropathy, and tumors. This study presents an overview of extracellular vesicles' and exosomes' pathogenic, diagnostic, and therapeutic values in inflammatory eye diseases, as well as existing and potential challenges.


Subject(s)
Cell-Derived Microparticles , Exosomes , Extracellular Vesicles , Eye Diseases , Humans , Extracellular Vesicles/metabolism , Exosomes/metabolism , Cell-Derived Microparticles/metabolism , Cell Communication/physiology , Eye Diseases/diagnosis , Eye Diseases/therapy , Eye Diseases/metabolism
17.
J Proteomics ; 270: 104738, 2023 01 06.
Article in English | MEDLINE | ID: mdl-36191803

ABSTRACT

Contact lens-related ocular surface complications occur more often in teenagers and young adults. The purpose of this study was to determine changes in tear proteome of young patients wearing glasses (GL), orthokeratology lenses (OK), and soft contact lenses (SCL). Twenty-two young subjects (10-26 years of age) who were established GL, OK, and SCL wearers were recruited. Proteomic data were collected using a data-independent acquisition-parallel accumulation serial fragmentation workflow. In total, 3406 protein groups were identified, the highest number of proteins identified in Schirmer strip tears to date. Eight protein groups showed higher abundance, and 11 protein groups showed lower abundance in the SCL group compared to the OK group. In addition, the abundance of 82 proteins significantly differed in children compared to young adult GL wearers, among which 67 proteins were higher, and 15 proteins were lower in children. These 82 proteins were involved in inflammation, immune, and glycoprotein metabolic biological processes. In summary, this work identified over 3000 proteins in Schirmer Strip tears. The results indicated that tear proteomes were altered by orthokeratology and soft contact wear and age, which warrants further larger-scale study on the ocular surface responses of teenagers and young adults separately to contact lens wear. SIGNIFICANCE: In this work, we examined the tear proteomes of young patients wearing glasses, orthokeratology lenses, and soft contact lenses using a data-independent acquisition-parallel accumulation serial fragmentation (diaPASEF) workflow and identified 3406 protein groups in Schirmer strip tears. Nineteen protein groups showed significant abundance changes between orthokeratology and soft contact lens wearers. Moreover, eighty-two protein groups significantly differed in abundance in children and young adult glasses wearers. As a pilot study, this work provides a deep coverage of tear proteome and suggests the need to investigate ocular responses to contact lens wear separately for children and young adults.


Subject(s)
Contact Lenses, Hydrophilic , Eye Diseases , Young Adult , Adolescent , Child , Humans , Proteome/metabolism , Proteomics , Pilot Projects , Tears/metabolism , Eye Diseases/metabolism
18.
Cells ; 11(23)2022 Dec 04.
Article in English | MEDLINE | ID: mdl-36497183

ABSTRACT

A layer of proteoglycans and glycoproteins known as glycocalyx covers the surface of the trabecular meshwork (TM), juxtacanalicular tissue (JCT), and Schlemm's canal (SC) inner wall of the conventional aqueous outflow pathway in the eye. This has been shown to play a role in the mechanotransduction of fluid shear stress and in the regulation of the outflow resistance. The outflow resistance in the conventional outflow pathway is the main determinant of the intraocular pressure (IOP) through an active, two-way, fluid-structure interaction coupling between the outflow tissues and aqueous humor. A 3D microstructural finite element (FE) model of a healthy human eye TM/JCT/SC complex with interspersed aqueous humor was constructed. A very thin charged double layer that represents the endothelial glycocalyx layer covered the surface of the elastic outflow tissues. The aqueous humor was modeled as electroosmotic flow that is charged when it is in contact with the outflow tissues. The electrical-fluid-structure interaction (EFSI) method was used to couple the charged double layer (glycocalyx), fluid (aqueous humor), and solid (outflow tissues). When the IOP was elevated to 15 mmHg, the maximum aqueous humor velocity in the EFSI model was decreased by 2.35 mm/s (9%) compared to the fluid-structure interaction (FSI) model. The charge or electricity in the living human conventional outflow pathway generated by the charged endothelial glycocalyx layer plays a minor biomechanical role in the resultant stresses and strains as well as the hydrodynamics of the aqueous humor.


Subject(s)
Eye Diseases , Mechanotransduction, Cellular , Humans , Trabecular Meshwork/metabolism , Aqueous Humor/metabolism , Intraocular Pressure , Glycocalyx , Eye Diseases/metabolism
19.
IUBMB Life ; 74(12): 1264-1272, 2022 12.
Article in English | MEDLINE | ID: mdl-36308309

ABSTRACT

Extracellular vesicles (EVs) are small packages that are released by almost all types of cells. While the role of EVs in pathogenesis of certain diseases such as cancer is well established, EVs role in ocular health and disease is still at early stages of investigation. Given the significant role of EVs in pathological development and progression of diseases such as cancer, EVs present a similar opportunity for investigation in ocular pathophysiology. Studies have shown the presence of EVs in fluids from the ocular environment have close links with ocular health and disease. Hence, the cargo carried in EVs from ocular fluids can be used for monitoring disease phenotypes or therapeutic outcomes in eye-related disorders. Furthermore, in recent times EVs have increasingly gained attention as therapeutics and drug-delivery vehicles for treatment of eye diseases. There is a close relationship between EVs and mitochondria functioning with mitochondria dysfunction leading to a significant number of ophthalmic disorders. This review discusses the current knowledge of EVs in visual systems with a special focus on eye diseases resulting from dysfunctional mitochondria.


Subject(s)
Extracellular Vesicles , Eye Diseases , Mitochondrial Diseases , Neoplasms , Humans , Extracellular Vesicles/metabolism , Drug Delivery Systems , Neoplasms/metabolism , Eye Diseases/metabolism , Mitochondria
20.
Cells ; 11(15)2022 07 29.
Article in English | MEDLINE | ID: mdl-35954181

ABSTRACT

The TGF-ß signaling pathway plays a crucial role in several key aspects of development and tissue homeostasis. TGF-ß ligands and their mediators have been shown to be important regulators of ocular physiology and their dysregulation has been described in several eye pathologies. TGF-ß signaling participates in regulating several key developmental processes in the eye, including angiogenesis and neurogenesis. Inadequate TGF-ß signaling has been associated with defective angiogenesis, vascular barrier function, unfavorable inflammatory responses, and tissue fibrosis. In addition, experimental models of corneal neovascularization, diabetic retinopathy, proliferative vitreoretinopathy, glaucoma, or corneal injury suggest that aberrant TGF-ß signaling may contribute to the pathological features of these conditions, showing the potential of modulating TGF-ß signaling to treat eye diseases. This review highlights the key roles of TGF-ß family members in ocular physiology and in eye diseases, and reviews approaches targeting the TGF-ß signaling as potential treatment options.


Subject(s)
Eye/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Diabetic Retinopathy/metabolism , Eye Diseases/metabolism , Homeostasis , Humans , Neovascularization, Pathologic
SELECTION OF CITATIONS
SEARCH DETAIL
...