Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Exp Eye Res ; : 110100, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39299675

RESUMEN

Diabetes mellitus (DM) is a common metabolic disease associated with severe macrovascular and microvascular complications that influence nearly every tissue in the body, including the anterior and posterior segments of the eye. In the cornea, DM is associated with recurrent epithelial erosion and reduced wound-healing capacity, which increases the risk of corneal scarring. We previously developed a co-culture model of the cornea consisting of immortalized human corneal epithelial cells (hCE-TJ) overlaying a self-assembled stromal layer generated by human corneal fibroblasts (hCFs) over a 4-week period. In this study, we investigated epithelial-stromal constructs generated from hCFs derived from subjects with Type 1 (T1DM) or 2 diabetes (T2DM) compared to controls. We found that T2DM constructs exhibited a disrupted epithelium and a thicker, stratified stromal layer compared to controls or T1DM. Both T1DM and T2DM stromal constructs expressed lower expression of thrombospondin-1 in isolated extracellular vesicles (EVs) compared to controls with no significant difference observed in the presence of epithelial cells, suggesting that reduced provisional matrix secretion in the corneal stroma may be a factor that promotes delayed corneal wound healing in diabetes. The tetraspanins are established extracellular vesicle (EV) markers and include CD63, CD81, and CD9, and were highly expressed by EVs in all three cell types. Control corneal stromal fibroblasts produced more and larger EVs when compared to T1DM and T2DM hCF-derived EVs, supporting a role for altered cell-cell communication in the context of DM. Further characterization of EVs and their cargo is expected to aid in the development of targeted treatments to improve corneal wound healing.

2.
Transl Vis Sci Technol ; 13(6): 12, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38888287

RESUMEN

Purpose: Recombinant human nerve growth factor (rhNGF; cenegermin-bkbj, OXERVATE) is the first and only U.S. Food and Drug Administration-approved treatment for moderate to severe neurotrophic keratopathy. The aim of this study was to determine the feasibility of incorporating a version of rhNGF in a mucoadhesive hydrogel capable of sustained drug release to the ocular surface. Methods: Hydrogels loaded with rhNGF were synthesized by conjugating chitosan with azidobenzoic acid (Az-Ch), adding rhNGF, and exposing the solution to ultraviolet (UV) radiation to induce photocrosslinking. Az-Ch hydrogels were evaluated for physical properties and rhNGF release profiles. Cytocompatbility of Az-Ch was assessed using immortalized human corneal limbal epithelial (HCLE) cells. TF1 erythroleukemic cell proliferation and HCLE cell proliferation and migration were used to assess the bioactivity of rhNGF released from Az-Ch hydrogels. Results: Az-Ch formed hydrogels in <10 seconds of UV exposure and demonstrated high optical transparency (75-85 T%). Az-Ch hydrogels exhibited good cytocompatibility with no demonstratable effect on HCLE cell morphology or viability. rhNGF was released gradually over 24 hours from Az-Ch hydrogels and retained its ability to induce TF1 cell proliferation. No significant difference was observed between rhNGF released from Az-Ch and freshly prepared rhNGF solutions on HCLE cell proliferation or percent wound closure after 12 hours; however, both were significantly better than control (P < 0.01). Conclusions: rhNGF-loaded Az-Ch hydrogels exhibited favorable physical, optical, and drug-release properties, as well as retained drug bioactivity. This drug delivery system has the potential to be further developed for in vivo and translational clinical applications. Translational Relevance: Az-Ch hydrogels may be used to enhance rhNGF therapy in patients with NK.


Asunto(s)
Proliferación Celular , Quitosano , Hidrogeles , Factor de Crecimiento Nervioso , Factor de Crecimiento Nervioso/farmacología , Factor de Crecimiento Nervioso/química , Factor de Crecimiento Nervioso/administración & dosificación , Humanos , Quitosano/química , Quitosano/farmacología , Hidrogeles/química , Hidrogeles/farmacología , Hidrogeles/síntesis química , Proliferación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Rayos Ultravioleta , Reactivos de Enlaces Cruzados/química , Limbo de la Córnea/efectos de los fármacos , Limbo de la Córnea/citología , Proteínas Recombinantes/química , Sistemas de Liberación de Medicamentos/métodos
3.
Int J Mol Sci ; 23(11)2022 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-35682600

RESUMEN

Communication between cells and the microenvironment is a complex, yet crucial, element in the development and progression of varied physiological and pathological processes. Accumulating evidence in different disease models highlights roles of extracellular vesicles (EVs), either in modulating cell signaling paracrine mechanism(s) or harnessing their therapeutic moiety. Of interest, the human cornea functions as a refractive and transparent barrier that protects the intraocular elements from the external environment. Corneal trauma at the ocular surface may lead to diminished corneal clarity and detrimental effects on visual acuity. The aberrant activation of corneal stromal cells, which leads to myofibroblast differentiation and a disorganized extracellular matrix is a central biological process that may result in corneal fibrosis/scarring. In recent years, understanding the pathological and therapeutic EV mechanism(s) of action in the context of corneal biology has been a topic of increasing interest. In this review, we describe the clinical relevance of corneal fibrosis/scarring and how corneal stromal cells contribute to wound repair and their generation of the stromal haze. Furthermore, we will delve into EV characterization, their subtypes, and the pathological and therapeutic roles they play in corneal scarring/fibrosis.


Asunto(s)
Enfermedades de la Córnea , Lesiones de la Cornea , Vesículas Extracelulares , Cicatriz/patología , Córnea/metabolismo , Enfermedades de la Córnea/etiología , Enfermedades de la Córnea/patología , Lesiones de la Cornea/metabolismo , Vesículas Extracelulares/metabolismo , Fibrosis , Humanos , Cicatrización de Heridas/fisiología
4.
Int J Mol Sci ; 23(6)2022 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-35328555

RESUMEN

Corneal epithelial wound healing is a multifaceted process that encompasses cell proliferation, migration, and communication from the corneal stroma. Upon corneal injury, bidirectional crosstalk between the epithelium and stroma via extracellular vesicles (EVs) has been reported. However, the mechanisms by which the EVs from human corneal keratocytes (HCKs), fibroblasts (HCFs), and/or myofibroblasts (HCMs) exert their effects on the corneal epithelium remain unclear. In this study, HCK-, HCF-, and HCM-EVs were isolated and characterized, and human corneal epithelial (HCE) cell migration was assessed in a scratch assay following PKH26-labeled HCK-, HCF-, or HCM-EV treatment. HCE cells proliferative and apoptotic activity following EV treatment was assessed. HCF-/HCM-EVs were enriched for CD63, CD81, ITGAV, and THBS1 compared to HCK-EV. All EVs were negative for GM130 and showed minimal differences in biophysical properties. At the proteomic level, we showed HCM-EV with a log >two-fold change in CXCL6, CXCL12, MMP1, and MMP2 expression compared to HCK-/HCF-EVs; these proteins are associated with cellular movement pathways. Upon HCM-EV treatment, HCE cell migration, velocity, and proliferation were significantly increased compared to HCK-/HCF-EVs. This study concludes that the HCM-EV protein cargo influences HCE cell migration and proliferation, and understanding these elements may provide a novel therapeutic avenue for corneal wound healing.


Asunto(s)
Lesiones de la Cornea , Epitelio Corneal , Vesículas Extracelulares , Movimiento Celular , Lesiones de la Cornea/metabolismo , Células Epiteliales/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Miofibroblastos/metabolismo , Proteómica
5.
Biomolecules ; 11(11)2021 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-34827680

RESUMEN

Corneal fibrosis (or scarring) occurs in response to ocular trauma or infection, and by reducing corneal transparency, it can lead to visual impairment and blindness. Studies highlight important roles for transforming growth factor (TGF)-ß1 and -ß3 as modulators in corneal wound healing and fibrosis, leading to increased extracellular matrix (ECM) components and expression of α-smooth muscle actin (αSMA), a myofibroblast marker. In this study, human corneal fibroblasts (hCF) were cultured as a monolayer culture (2D) or on poly-transwell membranes to generate corneal stromal constructs (3D) that were treated with TGF-ß1, TGF-ß3, or TGF-ß1 + FAK inhibitor (FAKi). Results show that hCF 3D constructs treated with TGF-ß1 or TGF-ß3 impart distinct effects on genes involved in wound healing and fibrosis-ITGAV, ITGB1, SRC and ACTA2. Notably, in the 3D construct model, TGF-ß1 enhanced αSMA and focal adhesion kinase (FAK) protein expression, whereas TGF-ß3 did not. In addition, in both the hCF 2D cell and 3D construct models, we found that TGF-ß1 + FAKi attenuated TGF-ß1-mediated myofibroblast differentiation, as shown by abrogated αSMA expression. This study concludes that FAK signaling is important for the onset of TGF-ß1-mediated myofibroblast differentiation, and FAK inhibition may provide a novel beneficial therapeutic avenue to reduce corneal scarring.


Asunto(s)
Fibroblastos , Factor de Crecimiento Transformador beta1 , Diferenciación Celular , Humanos , Miofibroblastos
6.
Anal Cell Pathol (Amst) ; 2021: 4793338, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34336553

RESUMEN

Epithelial wound healing is essential to repair the corneal barrier function after injury and requires coordinated epithelial sheet movement over the wounded region. The presence and role of pannexin1 on multilayered epithelial sheet migration was examined in unwounded and wounded corneal epithelium from C57BL/6J (B6) control and diet-induced obese (DiO) mice, a pretype 2 diabetic model. We hypothesize that pannexin1 is dysregulated, and the interaction of two ion-channel proteins (P2X7 and pannexin1) is altered in pretype 2 diabetic tissue. Pannexin1 was found to be present along cell borders in unwounded tissue, and no significant difference was observed between DiO and B6 control. However, an epithelial debridement induced a striking difference in pannexin1 localization. The B6 control epithelium displayed intense staining near the leading edge, which is the region where calcium mobilization was detected, whereas the staining in the DiO corneal epithelium was diffuse and lacked distinct gradation in intensity back from the leading edge. Cells distal to the wound in the DiO tissue were irregular in shape, and the morphology was similar to that of epithelium inhibited with 10Panx, a pannexin1 inhibitor. Pannexin1 inhibition reduced mobilization of calcium between cells near the leading edge, and MATLAB scripts revealed a reduction in cell-cell communication that was also detected in cultured cells. Proximity ligation was performed to determine if P2X7 and pannexin1 interaction was a necessary component of motility and communication. While there was no significant difference in the interaction in unwounded DiO and B6 control corneal epithelium, there was significantly less interaction in the wounded DiO corneas both near the wound and back from the edge. The results demonstrate that pannexin1 contributes to the healing response, and P2X7 and pannexin1 coordination may be a required component of cell-cell communication and an underlying reason for the lack of pathologic tissue migration.


Asunto(s)
Diabetes Mellitus , Epitelio Corneal , Animales , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Epitelio Corneal/metabolismo , Epitelio Corneal/patología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/fisiología , Cicatrización de Heridas/fisiología
7.
Anal Cell Pathol (Amst) ; 2021: 9983900, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34336556

RESUMEN

Extracellular vesicles (EVs) are phospholipid bilayer-bound particles secreted by cells that have been found to be important in mediating cell-cell communication, signal transduction, and extracellular matrix remodeling. Their role in both physiological and pathological processes has been established in different tissues throughout the human body. The human cornea functions as a transparent and refractive barrier that protects the intraocular elements from the external environment. Injury, infection, or disease may cause the loss of corneal clarity by altering extracellular matrix organization within the stroma that may lead to detrimental effects on visual acuity. Over the years, numerous studies have identified many of the growth factors (e.g., transforming growth factor-ß1, thrombospondin-1, and platelet-derived growth factor) important in corneal wound healing and scarring. However, the functional role of bound factors encapsulated in EVs in the context of corneal biology is less defined. In this review, we describe the discovery and characterization of EVs in the cornea. We focus on EV-matrix interactions, potential functions during corneal wound healing, and the bioactivity of mesenchymal stem cell-derived EVs. We also discuss the development of EVs as stable, drug-loaded therapeutics for ocular applications.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas , Comunicación Celular , Córnea/metabolismo , Córnea/patología , Vesículas Extracelulares/metabolismo , Humanos , Cicatrización de Heridas
8.
Curr Protoc Cell Biol ; 89(1): e114, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32986311

RESUMEN

Science and medicine have become increasingly "human-centric" over the years. A growing shift away from the use of animals in basic research has led to the development of sophisticated in vitro models of various tissues utilizing human-derived cells to study physiology and disease. The human cornea has likewise been modeled in vitro using primary cells derived from corneas obtained from cadavers or post-transplantation. By utilizing a cell's intrinsic ability to maintain its tissue phenotype in a pre-designed microenvironment containing the required growth factors, physiological temperature, and humidity, tissue-engineered corneas can be grown and maintained in culture for relatively long periods of time on the scale of weeks to months. Due to its transparency and avascularity, the cornea is an optimal tissue for studies of extracellular matrix and cell-cell interactions, toxicology and permeability of drugs, and underlying mechanisms of scarring and tissue regeneration. This paper describes methods for the cultivation of corneal keratocytes, fibroblasts, epithelial, and endothelial cells for in vitro applications. We also provide detailed, step-by-step protocols for assembling and culturing 3D constructs of the corneal stroma, epithelial- and endothelial-stromal co-cultures and isolation of extracellular vesicles. © 2020 Wiley Periodicals LLC. Basic Protocol 1: Isolating and culturing human corneal keratocytes and fibroblasts Basic Protocol 2: Isolating and culturing human corneal epithelial cells Basic Protocol 3: Isolating and culturing human corneal endothelial cells Basic Protocol 4: 3D corneal stromal construct assembly Basic Protocol 5: 3D corneal epithelial-stromal construct assembly Basic Protocol 6: 3D corneal endothelial-stromal construct assembly Basic Protocol 7: Isolating extracellular vesicles from corneal cell conditioned medium Support Protocol: Cryopreserving human corneal fibroblasts, corneal epithelial cells, and corneal endothelial cells.


Asunto(s)
Comunicación Celular , Córnea/citología , Técnicas Citológicas/métodos , Células Endoteliales/citología , Separación Celular , Células Cultivadas , Técnicas de Cocultivo , Sustancia Propia/citología , Criopreservación , Medios de Cultivo Condicionados/farmacología , Células Epiteliales/citología , Fibroblastos/citología , Humanos , Uniones Estrechas/metabolismo
9.
Exp Eye Res ; 197: 108127, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32619578

RESUMEN

The cornea is an avascular, transparent ocular tissue that serves as a refractive and protective structure for the eye. Over 90% of the cornea is composed of a collagenous-rich extracellular matrix within the stroma with the other 10% composed by the corneal epithelium and endothelium layers and their corresponding supporting collagen layers (e.g., Bowman's and Descemet's membranes) at the anterior and posterior cornea, respectively. Due to its prominent role in corneal structure, tissue engineering approaches to model the human cornea in vitro have focused heavily on the cellular and functional properties of the corneal stroma. In this review, we discuss model development in the context of culture dimensionality (e.g., 2-dimensional versus 3-dimensional) and expand on the optical, biomechanical, and cellular functions promoted by the culture microenvironment. We describe current methods to model the human cornea with focus on organotypic approaches, compressed collagen, bioprinting, and self-assembled stromal models. We also expand on co-culture applications with the inclusion of relevant corneal cell types, such as epithelial, stromal keratocyte or fibroblast, endothelial, and neuronal cells. Further advancements in corneal tissue model development will markedly improve our current understanding of corneal wound healing and regeneration.


Asunto(s)
Bioimpresión/métodos , Córnea/diagnóstico por imagen , Enfermedades de la Córnea/cirugía , Imagenología Tridimensional/métodos , Ingeniería de Tejidos/métodos , Células Cultivadas , Córnea/cirugía , Enfermedades de la Córnea/diagnóstico , Humanos
10.
Cells ; 9(5)2020 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-32357574

RESUMEN

The corneal epithelium mediates the initial response to injury of the ocular surface and secretes a number of profibrotic factors that promote corneal scar development within the stroma. Previous studies have shown that corneal epithelial cells also secrete small extracellular vesicles (EVs) in response to corneal wounding. In this paper, we hypothesized that EVs released from corneal epithelial cells in vitro contain protein cargo that promotes myofibroblast differentiation, the key cell responsible for scar development. We focused on the interplay between corneal epithelial-derived EVs and the stroma to determine if the corneal fibroblast phenotype, contraction, proliferation, or migration were promoted following vesicle uptake by corneal fibroblasts. Our results showed an increase in myofibroblast differentiation based on α-smooth muscle actin expression and elevated contractility following EV treatment compared to controls. Furthermore, we characterized the contents of epithelial cell-derived EVs using proteomic analysis and identified the presence of provisional matrix proteins, fibronectin and thrombospondin-1, as the dominant encapsulated protein cargo secreted by corneal epithelial cells in vitro. Proteins associated with the regulation of protein translation were also abundant in EVs. This paper reveals a novel role and function of EVs secreted by the corneal epithelium that may contribute to corneal scarring.


Asunto(s)
Epitelio Corneal/metabolismo , Vesículas Extracelulares/fisiología , Miofibroblastos/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Córnea/fisiología , Células Epiteliales/metabolismo , Vesículas Extracelulares/metabolismo , Fibroblastos/metabolismo , Fibronectinas , Humanos , Cultivo Primario de Células , Proteómica , Cicatrización de Heridas
11.
Anat Rec (Hoboken) ; 303(6): 1727-1734, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31136100

RESUMEN

One question that has intrigued cell biologists for many years is, "How do cells interact to influence one another's activity?" The discovery of extracellular vesicles (EVs) and the fact that they carry cargo, which directs cells to undergo changes in morphology and gene expression, has revolutionized this field of research. Little is known regarding the role of EVs in the cornea; however, we have demonstrated that EVs isolated from corneal epithelial cells direct corneal keratocytes to initiate fibrosis. Intriguingly, our data suggest that EVs do not penetrate epithelial basement membrane (BM), perhaps providing a mechanism explaining the importance of BM in the lack of scarring in scrape wounds. Since over 100-million people worldwide suffer from visual impairment as a result of corneal scarring, the role of EVs may be vital to understanding the mechanisms of wound repair. Therefore, we investigated EVs in ex vivo and in vivo-like three-dimensional cultures of human corneal cells using transmission electron microscopy. Some of the major findings were all three major cell types (epithelial, fibroblast, and endothelial cells) appear to release EVs, EVs can be identified using TEM, and EVs appeared to be involved in cell-cell communication. Interestingly, while our previous publication suggests that EVs do not penetrate the epithelial BM, it appears that EVs penetrate the much thicker endothelial BM (Descemet's membrane). These findings indicate the huge potential of EV research in the cornea and wound healing, and suggest that during homeostasis the endothelium and stromal cells are in communication. Anat Rec, 2019. © 2019 The Authors. The Anatomical Record published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.


Asunto(s)
Comunicación Celular/fisiología , Córnea/citología , Vesículas Extracelulares/metabolismo , Animales , Membrana Basal/metabolismo , Células Cultivadas , Córnea/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Conejos
12.
Eye (Lond) ; 34(2): 271-278, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31831879

RESUMEN

Corneal fibrosis develops in response to injury, infection, postsurgical complications, or underlying systemic disease that disrupts the homeostasis of the tissue leading to irregular extracellular matrix deposition within the stroma. The mechanisms that regulate corneal scarring are focused heavily on the canonical transforming growth factor-ß pathway and relevant activators, and their role in promoting myofibroblast differentiation. In this paper, we discuss the biochemical pathways involved in corneal fibrosis in the context of different injury models-epithelial debridement, superficial keratectomy, and penetrating incision. We elaborate on the interplay of the major pro-fibrotic factors involved in corneal scar development (e.g., transforming growth factor-ß1, thrombospondin-1, and ανß6), and explore a novel role for extracellular vesicles secreted by the wounded epithelium and the importance of the basement membrane.


Asunto(s)
Lesiones de la Cornea , Vesículas Extracelulares , Biología , Fibrosis , Humanos , Integrinas , Miofibroblastos/patología
13.
Bioengineering (Basel) ; 6(4)2019 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-31817298

RESUMEN

Cell-cell communication plays a fundamental role in mediating corneal wound healing following injury or infection. Depending on the severity of the wound, regeneration of the cornea and the propensity for scar development are influenced by the acute resolution of the pro-fibrotic response mediated by closure of the wound via cellular and tissue contraction. Damage of the corneal epithelium, basement membrane, and anterior stroma following a superficial keratectomy is known to lead to significant provisional matrix deposition, including secretion of fibronectin and thrombospondin-1, as well as development of a corneal scar. In addition, corneal wounding has previously been shown to promote release of extracellular vesicles from the corneal epithelium, which, in addition to soluble factors, may play a role in promoting tissue regeneration. In this study, we report the development and characterization of a co-culture system of human corneal epithelial cells and corneal stromal fibroblasts cultured for 4 weeks to allow extracellular matrix deposition and tissue maturation. The secretion of provisional matrix components, as well as small and large extracellular vesicles, was apparent within the constructs, suggesting cell-cell communication between epithelial and stromal cell populations. Laminin-1ß was highly expressed by the corneal epithelial layer with the presence of notable patches of basement membrane identified by transmission electron microscopy. Interestingly, we identified expression of collagen type III, fibronectin, and thrombospondin-1 along the epithelial-stromal interface similar to observations seen in vivo following a keratectomy, as well as expression of the myofibroblast marker, α-smooth muscle actin, within the stroma. Our results suggest that this corneal epithelial-stromal model may be useful in the study of the biochemical phenomena that occur during corneal wound healing.

14.
Sci Rep ; 9(1): 8079, 2019 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-31147562

RESUMEN

We previously demonstrated that inhibition of epidermal growth factor receptor (EGFR) slowed corneal epithelial migration. Here we examine the effect of EGF on transforming growth factor-beta receptor II (TGF-ßRII) in a corneal wound-healing model and primary human corneal epithelial cells (pHCE). Corneal debridement wounds were made and allowed to heal ± Tyrphostin AG1478 (EGFR inhibitor), and assayed for EGFR activation and EGFR and TGF-ßRII localization. Primary HCE were treated with EGF ± U0126 (MEK inhibitor) and assayed for TGF-ßRII expression. EGFR activation was maximal 15 minutes after wounding and localized in the migrating epithelial cells. TGF-ßRII localization was also observed in the migrating epithelium and was reduced when EGFR was blocked. When pHCE were treated with EGF for 6 hours, the cells produced enhanced levels of TGF-ßRII, which was blocked by U0126. Downstream signaling pathways of MEK (p38MAPK and ERK1/2MAPK) were then examined, and TGF-ß1 and EGF were found to have differential effects on the phosphorylation of p38 and ERK1/2, with TGF-ß1 upregulating p-p38 but not pERK1/2 and EGF upregulating pERK1/2 but not p-p38. Taken together, these data indicate that EGF stimulates TGF-ßRII through ERK1/2 and EGFR signaling, suggesting interplay between EGF- and TGF-ß-signaling pathways during corneal wound repair.


Asunto(s)
Lesiones de la Cornea/patología , Factor de Crecimiento Epidérmico/metabolismo , Epitelio Corneal/fisiología , Receptor Tipo II de Factor de Crecimiento Transformador beta/metabolismo , Cicatrización de Heridas/fisiología , Animales , Butadienos/farmacología , Células Cultivadas , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Epitelio Corneal/citología , Epitelio Corneal/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Nitrilos/farmacología , Cultivo Primario de Células , Quinazolinas/farmacología , Ratas , Factor de Crecimiento Transformador beta1/metabolismo , Tirfostinos/farmacología , Cicatrización de Heridas/efectos de los fármacos
15.
Exp Eye Res ; 184: 183-191, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30980816

RESUMEN

Corneal endothelium is a cellular monolayer positioned on the Descemet's membrane at the anterior cornea, and it plays a critical role in maintaining corneal clarity. Our present study examines the feasibility of utilizing our 3-dimensional (3D) corneal stromal construct, which consists of human corneal fibroblasts (HCF) and their self-assembled matrix, to observe the development and maturation of human corneal endothelial cells (HCEndoCs) in a co-culture model. Three-dimensional HCF constructs were created by growing the HCFs on Transwell membranes in Eagles' minimum essential medium (EMEM) + 10% FBS + 0.5 mM Vitamin C (VitC) for about 4 weeks. HCEndoCs, either primary (pHCEndoC) or cell line (HCEndoCL), were either seeded in chamber slides, directly on the Transwell membranes, or on the 3D HCF constructs and cultivated for 5 days or 2 weeks. The HCEndoCs that were seeded directly on the Transwell membranes were exposed indirectly to HCF by culturing the HCF on the plate beneath the membrane. Cultures were examined for morphology and ultrastructure using light and transmission electron microscopy (TEM). In addition, indirect-immunofluorescence microscopy (IF) was used to examine tight junction formation (ZO-1), maturation (ALDH1A1), basement membrane formation (Laminin), cell proliferation (Ki67), cell death (caspase-3), and fibrotic response (CTGF). As expected, both pHCEndoCs and HCEndoCLs formed monolayers on the constructs; however, the morphology of the HCEndoCLs appeared to be similar to that seen in vivo, uniform and closely packed, whereas the pHCEndoCs remained elongated. The IF data showed that laminin localization was present in the HCEndoCs' cytoplasm as cell-cell contact increased, and when they were grown in the 3D co-culture, the beginnings of what appears to be a continuous DM-like structure was observed. In addition, in co-cultures, ALDH1A1-positive HCEndoCs were present, ZO-1 expression localized within the tight junctions, minimal numbers of HCEndoCs were Ki67-or Caspase-3-positive, and CTGF was positive in both the HCEndoCs cytoplasm and the matrix of the co-culture. Also, laminin localization was stimulated in HCEndoCs upon indirect stimuli secreted by HCF. The present data suggests our 3D co-culture model is useful for studying corneal endothelium maturation in vitro since the co-culture promotes new DM-like formation, HCEndoCs develop in vivo-like characteristics, and the fibrotic response is activated. Our current findings are applicable to understanding the implications of corneal endothelial injection therapy, such as if the abnormal DM has to be removed from the patient, the newly injected endothelial cells will seed onto the wound area and deposit a new DM-like membrane. However, caution should be observed and as much of the normal DM should be left intact since removal of the DM can cause a posterior stromal fibrotic response.


Asunto(s)
Endotelio Corneal/citología , Imagenología Tridimensional , Modelos Biológicos , Familia de Aldehído Deshidrogenasa 1/metabolismo , Diferenciación Celular , Células Cultivadas , Técnicas de Cocultivo , Queratocitos de la Córnea/citología , Queratocitos de la Córnea/metabolismo , Queratocitos de la Córnea/ultraestructura , Lámina Limitante Posterior/metabolismo , Endotelio Corneal/metabolismo , Endotelio Corneal/ultraestructura , Humanos , Antígeno Ki-67/metabolismo , Laminina/metabolismo , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Retinal-Deshidrogenasa/metabolismo , Uniones Estrechas/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
16.
Exp Eye Res ; 180: 23-28, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30500364

RESUMEN

We previously demonstrated that ß6 knockout mice showed impaired wound repair in corneal debridement and keratectomy wounds. In the current investigation, we continued our examination of integrin αvß6 in order to determine if it was required for the initiation of wound healing in a corneal wound model that normally heals in a fibrotic manner. A full-thickness corneal incision was made in C57BL/6 J wild type (WT) and C57BL/6-Itgb6 KO (ß6-/-) mice. The mice were observed at 3, 7, 14, and 28 days post-incision. The morphology of corneal restoration was observed in tissue sections stained with hemotoxilin and eosin (H&E). In addition, indirect-immunofluorescence (IF) was performed on sections and/or whole mounts to evaluate the immunolocalization of α-smooth muscle actin (SMA) and thrombospondin-1 (TSP-1). H&E staining revealed that the corneas in ß6-/- mice healed slower than those in WT mice, with an obvious delay in the restoration of the stromal matrix and epithelium. In sections at 3 and 7 days, SMA and TSP-1 were greatly reduced in the ß6-/- mice as compared to WT, but peaked at 28 days after incision. Whole mount SMA IF results were consistent with those from sections. Therefore, the initiation of fibrosis was inhibited by the lack of αvß6; however, there appeared to be an alternate mechanism that initiated fibrosis 7-14 days later. Localization of TSP-1 correlated with expression of SMA whether wound healing was delayed or initiated immediately after wounding.


Asunto(s)
Antígenos de Neoplasias/fisiología , Córnea/patología , Lesiones de la Cornea/fisiopatología , Lesiones Oculares Penetrantes/fisiopatología , Integrinas/fisiología , Cicatrización de Heridas/fisiología , Actinas/metabolismo , Animales , Lesiones de la Cornea/metabolismo , Desbridamiento , Modelos Animales de Enfermedad , Femenino , Fibrosis/patología , Técnica del Anticuerpo Fluorescente Indirecta , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trombospondina 1/metabolismo
17.
Invest Ophthalmol Vis Sci ; 59(8): 3511-3520, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-30025094

RESUMEN

Purpose: Transforming growth factor-beta (TGF-ß) isoform 1 (T1) is involved in corneal fibrotic wound healing by stimulating myofibroblast transformation and altering fibrotic gene expression. In this study, two specific inhibitors were used to dissect the relationship between myofibroblast generation and the TGF-ß/Smad- or TGF-ß/p38-signaling pathway in human corneal fibroblasts (HCF). Methods: In HCF, Trx-SARA (Smad-pathway inhibitor) was used to block the TGF-ß/Smad-signaling pathway, and the p38 inhibitor (p38inh, SB202190) was used to inhibit p38MAPK, thus blocking the TGF-ß/p38-signaling pathway. HCF ± Trx-SARA or Trx-GA (SARA control) were serum starved overnight in Eagle's minimum essential medium (EMEM) ± p38inh, grown in EMEM ± T1 ± p38inh for 24 hours, and then processed for indirect-immunofluorescence, Western blot, or quantitative real-time polymerase chain reaction to examine α-smooth muscle actin (αSMA) and other fibrotic genes, such as fibronectin, thrombospondin1, and type III collagen. In addition, the morphology and the effect of p38inh on myofibroblast phenotype after myofibroblast formation were examined. Results: We observed that Trx-SARA had little effect on αSMA expression, indicating that blocking the Smad pathway did not significantly inhibit myofibroblast formation. However, p38inh did significantly inhibit αSMA and other fibrotic genes, thus efficiently preventing the transition of HCFs to myofibroblasts. In addition, morphology changed and αSMA decreased in myofibroblasts exposed to p38inh medium, as compared with controls. Conclusions: HCF transition to myofibroblasts was mainly through the p38 pathway. Therefore, blocking the p38 pathway may be a potential therapeutic tool for human corneal fibrosis prevention/treatment, because it controls myofibroblast formation in human corneal cells, while leaving other functions of T1 unaffected.


Asunto(s)
Queratocitos de la Córnea/citología , Sistema de Señalización de MAP Quinasas/fisiología , Miofibroblastos/citología , Factor de Crecimiento Transformador beta/metabolismo , Actinas/genética , Western Blotting , Línea Celular , Transdiferenciación Celular/fisiología , Células Cultivadas , Queratocitos de la Córnea/metabolismo , Electroforesis en Gel de Poliacrilamida , Inhibidores Enzimáticos/farmacología , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Imidazoles/farmacología , Miofibroblastos/metabolismo , Piridinas/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Smad/metabolismo
18.
Exp Eye Res ; 170: 127-137, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29496505

RESUMEN

Deposition of matrix proteins during development and repair is critical to the transparency of the cornea. While many cells respond to a hypoxic state that can occur in a tumor, the cornea is exposed to hypoxia during development prior to eyelid opening and during the diurnal sleep cycle where oxygen levels can drop from 21% to 8%. In this study, we used 2 three-dimensional (3-D) models to examine how stromal cells respond to periods of acute hypoxic states. The first model, a stromal construct model, is a 3-D stroma-like construct that consists of human corneal fibroblasts (HCFs) stimulated by a stable form of ascorbate for 1, 2, and 4 weeks to self-assemble their own extracellular matrix. The second model, a corneal organ culture model, is a corneal wound-healing model, which consists of wounded adult rat corneas that were removed and placed in culture to heal. Both models were exposed to either normoxic or hypoxic conditions for varying time periods, and the expression and/or localization of matrix proteins was assessed. No significant changes were detected in Type V collagen, which is associated with Type I collagen fibrils; however, significant changes were detected in the expression of both the small leucine-rich repeating proteoglycans and the larger heparan sulfate proteoglycan, perlecan. Also, hypoxia decreased both the number of Cuprolinic blue-positive glycosaminoglycan chains along collagen fibrils and Sulfatase 1, which modulates the effect of heparan sulfate by removing the 6-O-sulfate groups. In the stromal construct model, alterations were seen in fibronectin, similar to those that occur in development and after injury. These changes in fibronectin after injury were accompanied by changes in proteoglycans. Together these findings indicate that acute hypoxic changes alter the physiology of the cornea, and these models will allow us to manipulate the conditions in the extracellular environment in order to study corneal development and trauma.


Asunto(s)
Queratocitos de la Córnea/fisiología , Sustancia Propia/citología , Proteínas de la Matriz Extracelular/metabolismo , Matriz Extracelular/metabolismo , Hipoxia/metabolismo , Cicatrización de Heridas/fisiología , Animales , Ácido Ascórbico/farmacología , Colágeno/genética , Colágeno/metabolismo , Sustancia Propia/ultraestructura , Proteínas de la Matriz Extracelular/genética , Técnica del Anticuerpo Fluorescente Indirecta , Glicosaminoglicanos/genética , Glicosaminoglicanos/metabolismo , Humanos , Microscopía Confocal , Modelos Biológicos , Técnicas de Cultivo de Órganos , Proteoglicanos/genética , Proteoglicanos/metabolismo , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa
19.
Methods Mol Biol ; 1627: 117-122, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28836198

RESUMEN

Corneal scarring is an obligatory consequence of stroma corneal injury and is a major cause of decreased visual quality and vision loss worldwide. There are currently no satisfactory intervention therapies for corneal fibrosis. In this chapter, we describe well-established in vivo corneal wound models to allow researchers to investigate epithelial and stromal responses to corneal injury.


Asunto(s)
Cicatriz/etiología , Cicatriz/patología , Enfermedades de la Córnea/etiología , Enfermedades de la Córnea/patología , Modelos Animales de Enfermedad , Animales , Cicatriz/diagnóstico , Córnea/patología , Enfermedades de la Córnea/diagnóstico , Epitelio Corneal/patología , Ratones , Microscopía Fluorescente , Lámpara de Hendidura
20.
Artículo en Inglés | MEDLINE | ID: mdl-28649665

RESUMEN

PURPOSE: Transforming growth factor-beta (TGF-ß) activates the canonical Smad pathway, which includes the Smad family of proteins and SARA (Smad Anchor for Receptor Activation) and other less understood pathways, including one involving p38MAPK. The goal of the current research was to determine if corneal epithelial cells and fibroblasts used the classical or alternative TGF-ß-signaling pathways. To examine this question, we made use of Trx-SARA, which inhibits native SARA, thus blocking the Smad pathway. METHODS: A human corneal epithelial cell line (HCE-TJ), and stromal fibroblasts (HCF) were infected with retroviruses (RTV) containing either Trx-SARA or Trx-GA (a control plasmid). The effect of Trx-SARA on thrombospondin-1 (TSP-1) expression in both cell types, p15ink4b expression in HCE-TJ, and cellular fibronectin (cFN) expression in HCF was determined. In addition, the effect of p38MAPK inhibitor on TSP-1 and p15ink4b were examined. RESULTS: In HCE-TJ with TGF-ß1, TSP-1-protein levels increased and peaked at 24 hours. Trx-SARA reduced TSP-1 expression in HCE-TJ, but had no effect on p15ink4b. With HCF, Trx-SARA failed to reduce TSP-1 expression; however, cFN expression decreased and proliferation was inhibited. By blocking the p38MAPK pathway, TSP-1 expression was reduced in HCF and p15ink4b expression was decreased in HCE-TJ. CONCLUSIONS: Surprisingly, TSP-1 was regulated through the Smad pathway in HCE-TJ and the p38MAPK pathway in HCF. The p38MAPK pathway also induced p15ink4b in HCE-TJ. Our results indicate that not all TGF-ß-target proteins require the Smad pathway, and it may be possible to block certain TGF-ß-target proteins without blocking the expression of all the TGF-ß-target proteins.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA