Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Adv Res ; 45: 141-155, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-35623612

RESUMO

INTRODUCTION: Corneal blindness due to scarring is treated with corneal transplantation. However, a global problem is the donor material shortage. Preclinical and clinical studies have shown that cell-based therapy using corneal stromal stem cells (CSSCs) suppresses corneal scarring, potentially mediated by specific microRNAs transported in extracellular vesicles (EVs). However, not every CSSC batch from donors achieves similar anti-scarring effects. OBJECTIVES: To examine miRNA profiles in EVs from human CSSCs showing "healing" versus "non-healing" effects on corneal scarring and to design a tool to select CSSCs with strong healing potency for clinical applications. METHODS: Small RNAs from CSSC-EVs were extracted for Nanostring nCounter Human miRNA v3 assay. MicroRNAs expressed > 20 folds in "healing" EVs (P < 0.05) were subject to enriched gene ontology (GO) term analysis. MiRNA groups with predictive regulation on inflammatory and fibrotic signalling were studied by mimic transfection to (1) mouse macrophages (RAW264.7) for M1 phenotype assay; (2) human corneal keratocytes for cytokine-induced fibrosis, and (3) human CSSCs for corneal scar prevention in vivo. The expression of miR-29a was screened in additional CSSC batches and the anti-scarring effect of cells was validated in mouse corneal wounds. RESULTS: Twenty-one miRNAs were significantly expressed in "healing" CSSC-EVs and 9 miRNA groups were predicted to associate with inflammatory and fibrotic responses, and tissue regeneration (P <10-6). Overexpression of miR-29a and 381-5p significantly prevented M1 phenotype transition in RAW264.7 cells after lipopolysaccharide treatment, suppressed transforming growth factor ß1-induced fibrosis marker expression in keratocytes, and reduced scarring after corneal injury. High miR-29a expression in EV fractions distinguished human CSSCs with strong healing potency, which inhibited corneal scarring in vivo. CONCLUSION: We characterized the anti-inflammatory and fibrotic roles of miR-29a and 381-5p in CSSCs, contributing to scar prevention. MiR-29a expression in EVs distinguished CSSCs with anti-scarring quality, identifying good quality cells for a scarless corneal healing.


Assuntos
Lesões da Córnea , MicroRNAs , Humanos , Animais , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Lesões da Córnea/terapia , Células-Tronco/metabolismo , Cicatriz , Fibrose , Terapia Baseada em Transplante de Células e Tecidos
2.
Int J Mol Sci ; 23(21)2022 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-36362184

RESUMO

In addition to their therapeutic potential in regenerative medicine, human corneal stromal stem cells (CSSCs) could serve as a powerful tool for drug discovery and development. Variations from different donors, their isolation method, and their limited life span in culture hinder the utility of primary human CSSCs. To address these limitations, this study aims to establish and characterize immortalized CSSC lines (imCSSC) generated from primary human CSSCs. Primary CSSCs (pCSSC), isolated from human adult corneoscleral tissue, were transduced with ectopic expression of hTERT, c-MYC, or the large T antigen of the Simian virus 40 (SV40T) to generate imCSSC. Cellular morphology, proliferation capacity, and expression of CSSCs specific surface markers were investigated in all cell lines, including TNFAIP6 gene expression levels in vitro, a known biomarker of in vivo anti-inflammatory efficacy. SV40T-overexpressing imCSSC successfully extended the lifespan of pCSSC while retaining a similar morphology, proliferative capacity, multilineage differentiation potential, and anti-inflammatory properties. The current study serves as a proof-of-concept that immortalization of CSSCs could enable a large-scale source of CSSC for use in regenerative medicine.


Assuntos
Substância Própria , Células Estromais , Adulto , Humanos , Diferenciação Celular/fisiologia , Linhagem Celular , Células-Tronco
3.
Eye Vis (Lond) ; 7(1): 52, 2020 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-33292650

RESUMO

BACKGROUND: Corneal stromal stem cells (CSSC) reduce corneal inflammation, prevent fibrotic scarring, and regenerate transparent stromal tissue in injured corneas. These effects rely on factors produced by CSSC to block the fibrotic gene expression. This study investigated the mechanism of the scar-free regeneration effect. METHODS: Primary human CSSC (hCSSC) from donor corneal rims were cultivated to passage 3 and co-cultured with mouse macrophage RAW264.7 cells induced to M1 pro-inflammatory phenotype by treatment with interferon-γ and lipopolysaccharides, or to M2 anti-inflammatory phenotype by interleukin-4, in a Transwell system. The time-course expression of human transforming growth factor ß3 (hTGFß3) and hTGFß1 were examined by immunofluorescence and qPCR. TGFß3 knockdown for > 70% in hCSSC [hCSSC-TGFß3(si)] was achieved by small interfering RNA transfection. Naïve CSSC and hCSSC-TGFß3(si) were transplanted in a fibrin gel to mouse corneas, respectively, after wounding by stromal ablation. Corneal clarity and the expression of mouse inflammatory and fibrosis genes were examined. RESULTS: hTGFß3 was upregulated by hCSSC when co-cultured with RAW cells under M1 condition. Transplantation of hCSSC to wounded mouse corneas showed significant upregulation of hTGFß3 at days 1 and 3 post-injury, along with the reduced expression of mouse inflammatory genes (CD80, C-X-C motif chemokine ligand 5, lipocalin 2, plasminogen activator urokinase receptor, pro-platelet basic protein, and secreted phosphoprotein 1). By day 14, hCSSC treatment significantly reduced the expression of fibrotic and scar tissue genes (fibronectin, hyaluronan synthase 2, Secreted protein acidic and cysteine rich, tenascin C, collagen 3a1 and α-smooth muscle actin), and the injured corneas remained clear. However, hCSSC-TGFß3(si) lost these anti-inflammatory and anti-scarring functions, and the wounded corneas showed intense scarring. CONCLUSION: This study has demonstrated that the corneal regenerative effect of hCSSC is mediated by TGFß3, inducing a scar-free tissue response.

4.
Exp Eye Res ; 200: 108270, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32979396

RESUMO

Corneal opacities affect vision for millions of individuals worldwide. Fibrotic scar tissues accumulate in reaction to inflammatory responses and remain permanently in corneal stroma, and conventionally correctable only by donor corneal transplantation. Numerous studies have explored innovative approaches to reverse corneal scarring through non-surgical means; however, existing mouse models limit these studies, due to the lack of visibility of scar tissue in mouse corneas with steep curvature. Here, we reported that corneal scarring was modelled using a transgenic mouse line, Tg(Col3a1-EGFP)DJ124Gsat, in which enhanced green fluorescence protein (EGFP) reporter expression was driven by the promoter of collagen 3a1 (COL3a1), a stromal fibrosis gene. Similar to wildtype, Col3a1-EGFP transgenic corneas developed opacities after wounding by alkali burn and mechanical ablation, respectively, as examined under stereomicroscopy and Spectral Domain optical coherent tomography. The time course induction of EGFP was aligned with Col3a1 upregulation and matched with the elevated expression of other fibrosis genes (α-smooth muscle actin, fibronectin and tenascin C). Measured by flow cytometry and enzyme-linked immunosorbent assay, increased number of EGFP expressing cells and fluorescent intensities were correlated to corneal thickening and scar volume. After treatment with human corneal stromal stem cells or their exosomes, EGFP expression was downregulated together with the reduction of scar volume and fibrosis gene expression. These results have demonstrated that the transgenic mouse line, Tg(Col3a1-EGFP)DJ124Gsat, can be a valuable tool for the detection of corneal fibrosis and scarring in vivo, and will be useful in monitoring the changes of corneal fibrosis over time.


Assuntos
Cicatriz/diagnóstico , Colágeno Tipo III/genética , Lesões da Córnea/diagnóstico , Substância Própria/patologia , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Animais , Cicatriz/genética , Cicatriz/metabolismo , Colágeno Tipo III/biossíntese , Lesões da Córnea/genética , Lesões da Córnea/metabolismo , Substância Própria/metabolismo , Ensaio de Imunoadsorção Enzimática , Proteínas de Fluorescência Verde/biossíntese , Humanos , Camundongos , Camundongos Transgênicos , RNA/genética
5.
Invest Ophthalmol Vis Sci ; 60(8): 3013-3023, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31310658

RESUMO

Purpose: Mesenchymal stem cells (MSCs) have been extensively studied for their capacity to enhance wound healing and represent a promising research field for generating cell therapies for corneal scars. In the present study, we investigated MSCs from different tissues and their potential to differentiate toward corneal keratocytes. Methods: Adipose-derived stem cells, bone marrow MSCs, umbilical cord stem cells, and corneal stromal stem cells (CSSCs) were characterized by their expression of surface markers CD105, CD90, and CD73, and their multilineage differentiation capacity into adipocytes, osteoblasts, and chondrocytes. MSCs were also evaluated for their potential to differentiate toward keratocytes, and for upregulation of the anti-inflammatory protein TNFα-stimulated gene-6 (TNFAIP6) after simulation by IFN-γ and TNF-α. Results: Keratocyte lineage induction was achieved in all MSCs as indicated by the upregulated expression of keratocyte markers, including keratocan, lumican, and carbohydrate sulfotransferase. TNFAIP6 response to inflammatory stimulation was observed only in CSSCs; increasing by 3-fold compared with the control (P < 0.05). Conclusions: Based on our findings, CSSCs appeared to have the greatest differentiation potential toward the keratocyte lineage and the greatest anti-inflammatory properties in vitro.


Assuntos
Moléculas de Adesão Celular/genética , Ceratócitos da Córnea/citologia , Regulação da Expressão Gênica , Células-Tronco Mesenquimais/citologia , Moléculas de Adesão Celular/biossíntese , Diferenciação Celular , Células Cultivadas , Ceratócitos da Córnea/metabolismo , Citometria de Fluxo , Humanos , Células-Tronco Mesenquimais/metabolismo , RNA/genética , Fator de Necrose Tumoral alfa
6.
Stem Cells Transl Med ; 8(11): 1192-1201, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31290598

RESUMO

Mesenchymal stem cells from corneal stromal stem cells (CSSC) prevent fibrotic scarring and stimulate regeneration of transparent stromal tissue after corneal wounding in mice. These effects rely on the ability of CSSC to block neutrophil infiltration into the damaged cornea. The current study investigated the hypothesis that tissue regeneration by CSSC is mediated by secreted extracellular vesicles (EVs). CSSC produced EVs 130-150 nm in diameter with surface proteins that include CD63, CD81, and CD9. EVs from CSSC reduced visual scarring in murine corneal wounds as effectively as did live cells, but EVs from human embryonic kidney (HEK)293T cells had no regenerative properties. CSSC EV treatment of wounds decreased expression of fibrotic genes Col3a1 and Acta2, blocked neutrophil infiltration, and restored normal tissue morphology. CSSC EVs labeled with carboxyfluorescein succinimidyl ester dye, rapidly fused with corneal epithelial and stromal cells in culture, transferring microRNA (miRNA) to the target cells. Knockdown of mRNA for Alix, a component of the endosomal sorting complex required for transport, using siRNA, resulted in an 85% reduction of miRNA in the secreted EVs. The EVs with reduced miRNA were ineffective at blocking corneal scarring. Furthermore, CSSC with reduced Alix expression also lost their regenerative function, suggesting EVs as an obligate component in the delivery of miRNA. The results of these studies support an essential role for extracellular vesicles in the process by which CSSC cells block scarring and initiate regeneration of transparent corneal tissue after wounding. EVs appear to serve as a delivery vehicle for miRNA, which affects the regenerative action. Stem Cells Translational Medicine 2019;8:1192-1201.


Assuntos
Doenças da Córnea/terapia , Vesículas Extracelulares/transplante , Fibrose/terapia , Inflamação/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , MicroRNAs/administração & dosagem , Animais , Feminino , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Cicatrização
7.
Sci Rep ; 8(1): 17294, 2018 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-30470798

RESUMO

Diabetes mellitus is a disease caused by innate or acquired insulin deficiency, resulting in altered glucose metabolism and high blood glucose levels. Chronic hyperglycemia is linked to development of several ocular pathologies affecting the anterior segment, including diabetic corneal neuropathy and keratopathy, neovascular glaucoma, edema, and cataracts leading to significant visual defects. Due to increasing disease prevalence, related medical care costs, and visual impairment resulting from diabetes, a need has arisen to devise alternative systems to study molecular mechanisms involved in disease onset and progression. In our current study, we applied a novel 3D in vitro model of the human cornea comprising of epithelial, stromal, and neuronal components cultured in silk scaffolds to study the pathological effects of hyperglycemia on development of diabetic corneal neuropathy. Specifically, exposure to sustained levels of high glucose, ranging from 35 mM to 45 mM, were applied to determine concentration-dependent effects on nerve morphology, length and density of axons, and expression of metabolic enzymes involved in glucose metabolism. By comparing these metrics to in vivo studies, we have developed a functional 3D in vitro model for diabetic corneal neuropathy as a means to investigate corneal pathophysiology resulting from prolonged exposure to hyperglycemia.


Assuntos
Córnea/fisiopatologia , Doenças da Córnea/patologia , Diabetes Mellitus/fisiopatologia , Neuropatias Diabéticas/patologia , Hiperglicemia/fisiopatologia , Modelos Biológicos , Doenças do Sistema Nervoso Periférico/patologia , Células Cultivadas , Córnea/inervação , Doenças da Córnea/etiologia , Complicações do Diabetes/etiologia , Complicações do Diabetes/patologia , Diabetes Mellitus/induzido quimicamente , Neuropatias Diabéticas/etiologia , Glucose/efeitos adversos , Humanos , Hiperglicemia/induzido quimicamente , Técnicas In Vitro , Doenças do Sistema Nervoso Periférico/etiologia , Edulcorantes/efeitos adversos
8.
Adv Healthc Mater ; 7(19): e1800488, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30091220

RESUMO

New in vitro tissue models to mimic in vivo conditions are needed to provide insight into mechanisms involved in peripheral pain responses, potential therapeutic strategies to address these responses, and to replace animal models for such indications. For example, the rabbit cornea Draize test has become the standard method used for decades to screen ophthalmic drug and consumer product toxicity. In vitro tissue models with functional innervation have the potential to replace in vivo animal testing and provide sophisticated bench tools to study ocular nociception and its amelioration. Herein, full thickness, innervated, 3D human corneal tissues are grown under physiologically relevant culture conditions to study nociceptive-related responses, by mimicking ocular environmental cues, including intraocular pressure (IOP) and tear flow (TF). Capsaicin, a chili pepper-derived irritant known to cause a burning sensation in mammalian tissues is utilized as a nociceptive stimulant to induce pain, while subsequent serum treatment is used to mimic healing. Pain mediators released upon capsaicin stimulation and cell regrowth after serum treatment are characterized to assess ocular responses in this new, innervated, human corneal tissue system for comparison of outcomes to established animal and related responses.


Assuntos
Córnea/patologia , Dor Nociceptiva/induzido quimicamente , Capsaicina/toxicidade , Células Cultivadas , Córnea/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Cicatrização/fisiologia
9.
Stem Cells Transl Med ; 7(6): 487-494, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29654654

RESUMO

Stem cells from human corneal stroma (CSSC) suppress corneal stromal scarring in a mouse wound-healing model and promote regeneration of native transparent tissue (PMID:25504883). This study investigated efficacy of compressed collagen gel (CCG) as a vehicle to deliver CSSC for corneal therapy. CSSC isolated from limbal stroma of human donor corneas were embedded in soluble rat-tendon collagen, gelled at 37°C, and partially dehydrated to a thickness of 100 µm by passive absorption. The CCG disks were dimensionally stable, easy to handle, and could be adhered securely to de-epithelialized mouse cornea with fibrin-based adhesive. CSSC in CCG maintained >80% viability for >1 week in culture media and could be cryopreserved in 20% fetal bovine serum-10%DMSO in liquid nitrogen. CCG containing as few as 500 CSSC effectively prevented visible scarring and suppressed expression of fibrotic Col3a1 mRNA. CSSC in CCG were more effective at blocking scarring on a per-cell basis than CSSC delivered directly in a fibrin gel as previously described. Collagen-embedded cells retained the ability to suppress corneal scarring after conventional cryopreservation. This study demonstrates use of a common biomaterial that can facilitate storage and handling of stem cells in a manner that may provide off-the-shelf delivery of stem cells as a therapy for corneal scarring. Stem Cells Translational Medicine 2018;7:487-494.


Assuntos
Cicatriz/terapia , Colágeno/química , Transplante de Células-Tronco , Células-Tronco/citologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Terapia Baseada em Transplante de Células e Tecidos , Córnea/citologia , Córnea/patologia , Criopreservação , Modelos Animais de Doenças , Feminino , Humanos , Hidrogéis/química , Hidrogéis/farmacologia , Camundongos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Engenharia Tecidual/métodos , Alicerces Teciduais
10.
J Tissue Eng Regen Med ; 12(1): 59-69, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-27863068

RESUMO

Blinding corneal scarring is predominately treated with allogeneic graft tissue; however, there is a worldwide shortage of donor tissue leaving millions in need of therapy. Human corneal stromal stem cells (CSSC) have been shown produce corneal tissue when cultured on nanofibre scaffolding, but this tissue cannot be readily separated from the scaffold. In this study, scaffold-free tissue engineering methods were used to generate biomimetic corneal stromal tissue constructs that can be transplanted in vivo without introducing the additional variables associated with exogenous scaffolding. CSSC were cultured on substrates with aligned microgrooves, which directed parallel cell alignment and matrix organization, similar to the organization of native corneal stromal lamella. CSSC produced sufficient matrix to allow manual separation of a tissue sheet from the grooved substrate. These constructs were cellular and collagenous tissue sheets, approximately 4 µm thick and contained extracellular matrix molecules typical of corneal tissue including collagen types I and V and keratocan. Similar to the native corneal stroma, the engineered corneal tissues contained long parallel collagen fibrils with uniform diameter. After being transplanted into mouse corneal stromal pockets, the engineered corneal stromal tissues became transparent, and the human CSSCs continued to express human corneal stromal matrix molecules. Both in vitro and in vivo, these scaffold-free engineered constructs emulated stromal lamellae of native corneal stromal tissues. Scaffold-free engineered corneal stromal constructs represent a novel, potentially autologous, cell-generated, biomaterial with the potential for treating corneal blindness. Copyright © 2016 John Wiley & Sons, Ltd.


Assuntos
Substância Própria/fisiologia , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Substância Própria/ultraestrutura , Matriz Extracelular/metabolismo , Humanos , Implantes Experimentais , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco/citologia
11.
J Tissue Eng Regen Med ; 12(1): 285-295, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28600807

RESUMO

With insufficient options to meet the clinical demand for cornea transplants, one emerging area of emphasis is on cornea tissue engineering. In the present study, the goal was to combine the corneal stroma and epithelium into one coculture system, to monitor both human corneal stromal stem cell (hCSSC) and human corneal epithelial cell (hCE) growth and differentiation into keratocytes and differentiated epithelium in these three-dimensional tissue systems in vitro. Coculture conditions were first optimized, including the medium, air-liquid interface culture, and surface topography and chemistry of biomaterial scaffold films based on silk protein. The silk was used as scaffolding for both stromal and epithelial tissue layers because it is cell compatible, can be surface patterned, and is optically clear. Next, the effects of proliferating and differentiating hCEs and hCSSCs were studied in this in vitro system, including the effects on cell proliferation, matrix formation by immunochemistry, and gene expression by quantitative reverse transcription-polymerase chain reaction. The incorporation of both cell types into the coculture system demonstrated more complete differentiation and growth for both cell types compared to the corneal stromal cells and corneal epithelial cells alone. Silk films for corneal epithelial culture were optimized to combine a 4.0-µm-scale surface pattern with bulk-loaded collagen type IV. Differentiation of each cell type was in evidence based on increased expression of corneal stroma and epithelial proteins and transcript levels after 6 weeks in coculture on the optimized silk scaffolds.


Assuntos
Técnicas de Cocultura/métodos , Substância Própria/citologia , Epitélio Corneano/citologia , Seda/farmacologia , Células-Tronco/citologia , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Humanos , Fenótipo , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Engenharia Tecidual
12.
PLoS One ; 12(3): e0171712, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28257425

RESUMO

Corneal scarring limits vision for millions of individuals worldwide. Corneal transplantation (keratoplasty) is the standard of care for corneal opacity; however, it bears the risk of graft rejection and infection and is not universally available. Stem cell therapy holds promise as an alternative to keratoplasty. Stem cells from human corneal stroma (CSSC) induce regeneration of transparent corneal tissue in a mouse wound-healing model. In this study we investigated the mechanism by which CSSC prevent deposition of fibrotic tissue. Infiltration by CD11b+/Ly6G+ neutrophils and myeloperoxidase expression were increased in corneas 24 hr after corneal wounding but were reduced in CSSC-treated wounds. Secretion of TSG-6, a protein known to regulate neutrophil migration, was up-regulated in CSSC in response to TNFα and as CSSC differentiate to keratocytes. In vivo, wounded mouse corneas treated with CSSC contained human TSG-6. Inhibition of neutrophil infiltration into cornea by CSSC was reversed when TSG-6 expression was knocked down using siRNA. Silencing of TSG-6 expression in CSSC reduced their ability to block scarring and the expression of mRNA for fibrosis-associated proteins collagen III, tenascin C, and smooth muscle actin in wounded corneas. Neutropenic mice exhibited a significant reduction in corneal scarring and fibrotic mRNA expression 2 weeks after wounding. These results support the conclusion that neutrophil infiltration is an essential event in the fibrotic response to corneal damage and that prevention of scarring by CSSC is mediated by secretion of TSG-6 by these cells.


Assuntos
Lesões da Córnea/terapia , Ceratócitos da Córnea/transplante , Substância Própria/transplante , Transplante de Células-Tronco , Animais , Moléculas de Adesão Celular/genética , Córnea/metabolismo , Córnea/fisiopatologia , Lesões da Córnea/fisiopatologia , Substância Própria/fisiopatologia , Transplante de Córnea , Rejeição de Enxerto/fisiopatologia , Humanos , Camundongos , Infiltração de Neutrófilos/genética , Regeneração/genética , Cicatrização
13.
PLoS One ; 12(1): e0169504, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28099503

RESUMO

The worldwide need for human cornea equivalents continues to grow. Few clinical options are limited to allogenic and synthetic material replacements. We hypothesized that tissue engineered human cornea systems based on mechanically robust, patterned, porous, thin, optically clear silk protein films, in combination with human corneal stromal stem cells (hCSSCs), would generate 3D functional corneal stroma tissue equivalents, in comparison to previously developed 2D approaches. Silk film contact guidance was used to control the alignment and distribution of hCSSCs on RGD-treated single porous silk films, which were then stacked in an orthogonally, multi-layered architecture and cultured for 9 weeks. These systems were compared similar systems generated with human corneal fibroblasts (hCFs). Both cell types were viable and preferentially aligned along the biomaterial patterns for up to 9 weeks in culture. H&E histological sections showed that the systems seeded with the hCSSCs displayed ECM production throughout the entire thickness of the constructs. In addition, the ECM proteins tested positive for keratocyte-specific tissue markers, including keratan sulfate, lumican, and keratocan. The quantification of hCSSC gene expression of keratocyte-tissue markers, including keratocan, lumican, human aldehyde dehydrogenase 3A1 (ALDH3A1), prostaglandin D2 synthase (PTDGS), and pyruvate dehydrogenase kinase, isozyme 4 (PDK4), within the 3D tissue systems demonstrated upregulation when compared to 2D single silk films and to the systems generated with the hCFs. Furthermore, the production of ECM from the hCSSC seeded systems and subsequent remodeling of the initial matrix significantly improved cohesiveness and mechanical performance of the constructs, while maintaining transparency after 9 weeks.


Assuntos
Substância Própria/citologia , Seda/química , Células-Tronco/citologia , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Sobrevivência Celular , Células Cultivadas , Ceratócitos da Córnea/fisiologia , Substância Própria/fisiologia , Dimetilpolisiloxanos/química , Fibroblastos/citologia , Regulação da Expressão Gênica , Humanos , Proteoglicanas/metabolismo , Células Estromais/citologia , Células Estromais/fisiologia
14.
Biomaterials ; 112: 1-9, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27741498

RESUMO

The interactions between corneal nerve, epithelium, and stroma are essential for maintaining a healthy cornea. Thus, corneal tissue models that more fully mimic the anatomy, mechanical properties and cellular components of corneal tissue would provide useful systems to study cellular interactions, corneal diseases and provide options for improved drug screening. Here a corneal tissue model was constructed to include the stroma, epithelium, and innervation. Thin silk protein film stacks served as the scaffolding to support the corneal epithelial and stromal layers, while a surrounding silk porous sponge supported neuronal growth. The neurons innervated the stromal and epithelial layers and improved function and viability of the tissues. An air-liquid interface environment of the corneal tissue was also mimicked in vitro, resulting in a positive impact on epithelial maturity. The inclusion of three cell types in co-culture at an air-liquid interface provides an important advance for the field of in vitro corneal tissue engineering, to permit improvements in the study of innervation and corneal tissue development, corneal disease, and tissue responses to environmental factors.


Assuntos
Córnea/citologia , Córnea/crescimento & desenvolvimento , Técnicas de Cultura de Órgãos/métodos , Impressão Tridimensional , Engenharia Tecidual/métodos , Órgãos Bioartificiais , Células Cultivadas , Técnicas de Cocultura/métodos , Córnea/inervação , Humanos , Neurônios/citologia , Neurônios/fisiologia , Seda/química , Células Estromais/citologia , Células Estromais/fisiologia , Engenharia Tecidual/instrumentação , Alicerces Teciduais
15.
Adv Healthc Mater ; 5(22): 2942-2950, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27723276

RESUMO

Basement membranes are protein-rich extracellular matrices (ECM) that are essential for epithelial and endothelial tissue structure and function. Aging and disease cause changes in the physical properties and ECM composition of basement membranes, which has spurred research to develop methods to repair and/or regenerate these tissues. An area of critical clinical need is the cornea, where failure of the endothelium leads to stromal edema and vision loss. Here, an engineered basement membrane (EBM) is developed that consists of a dense layer of collagen IV and/or laminin ≈5-10 nm thick, created using surface-initiated assembly, conformally attached to a collagen I film. These EBMs are used to engineer a corneal endothelium (CE) that mimics the structure of Descemet's membrane with a thin stromal layer, toward use as a graft for lamellar keratoplasty. Results show that bovine and human CE cells form confluent monolayers on the EBM, express ZO-1 at the cell-cell borders, and achieve a density of ≈1600 cells mm-2 for 28 and 14 d, respectively. These results demonstrate that the technique is capable of fabricating EBMs with structural and compositional properties that mimic native basement membranes and that EBM may be a suitable carrier for engineering transplant quality CE grafts.


Assuntos
Membrana Basal/citologia , Córnea/citologia , Endotélio Corneano/citologia , Regeneração/fisiologia , Adolescente , Adulto , Animais , Membrana Basal/metabolismo , Bovinos , Células Cultivadas , Colágeno Tipo I/metabolismo , Colágeno Tipo IV/metabolismo , Córnea/metabolismo , Lâmina Limitante Posterior/citologia , Lâmina Limitante Posterior/metabolismo , Endotélio Corneano/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Feminino , Humanos , Laminina/metabolismo , Masculino
16.
Sci Rep ; 6: 30987, 2016 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-27527833

RESUMO

Given the increasing emergence of antimicrobial resistant microbes and the near absent development of new antibiotic classes, innovative new therapeutic approaches to address this global problem are necessary. The use of predatory bacteria, bacteria that prey upon other bacteria, is gaining interest as an "out of the box" therapeutic treatment for multidrug resistant pathogenic bacterial infections. Before a new antimicrobial agent is used to treat infections, it must be tested for safety. The goal of this study was to test the tolerability of bacteria on the ocular surface using in vitro and in vivo models. Predatory bacteria Bdellovibrio bacteriovorus and Micavibrio aeruginosavorus were found to be non-toxic to human corneal stromal keratocytes in vitro; however, they did induce production of the proinflammatory chemokine IL-8 but not IL-1ß. Predatory bacteria did not induce inflammation on the ocular surface of rabbit eyes, with and without corneal epithelial abrasions. Unlike a standard of care antibiotic vancomycin, predatory bacteria did not inhibit corneal epithelial wound healing or increase clinical inflammatory signs in vivo. Together these data support the safety of predatory bacteria on the ocular surface, but future studies are warranted regarding the use predatory bacteria in deeper tissues of the eye.


Assuntos
Alphaproteobacteria/crescimento & desenvolvimento , Antibiose , Bdellovibrio bacteriovorus/crescimento & desenvolvimento , Olho/imunologia , Inflamação/imunologia , Cicatrização/fisiologia , Animais , Olho/microbiologia , Feminino , Humanos , Inflamação/microbiologia , Coelhos
17.
J Cataract Refract Surg ; 42(2): 302-9, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27026456

RESUMO

PURPOSE: To use a well-established organ culture model to investigate the effects of corneal stromal stem cells on the optical and biomechanical properties of corneal wounds after laser in situ keratomileusis (LASIK)-like flap creation. SETTING: School of Optometry and Vision Sciences, Cardiff University, Cardiff, Wales, United Kingdom. DESIGN: Experimental study. METHODS: The LASIK-like flaps were produced in sheep corneas. The flap beds were treated with corneal stromal stem cells and were then replaced and allowed to heal for different periods of up to 3 weeks in organ culture. The optical transmission of the cornea, the force required to detach the flap, and the presence of myofibroblasts near the flap bed were measured. RESULTS: Corneal stromal stem cell-treated flap beds were statistically significantly more transparent after 3 weeks in culture than the untreated controls. At 3 weeks, the mean force necessary to detach the flap was more than twice the force required for the respective control samples. Concurrently, there were 44% activated cells immediately below the flap margin of the controls compared with 29% in the same region of the corneal stromal stem cell-treated flaps. CONCLUSIONS: In this system, the presence of corneal stromal stem cells at the wound margin significantly increased the adherence of LASIK-like flaps while maintaining corneal transparency. It is postulated that this is achieved by the deposition of extracellular connective tissue similar to that found in the normal cornea and by the paucity of activated keratocytes (myofibroblasts), which are known to scatter a significant amount of the incident light. FINANCIAL DISCLOSURE: No author has a financial or proprietary interest in any material or method mentioned.


Assuntos
Córnea/cirurgia , Substância Própria/citologia , Ceratomileuse Assistida por Excimer Laser In Situ , Lasers de Excimer , Transplante de Células-Tronco , Células-Tronco/fisiologia , Cicatrização/fisiologia , Actinas/metabolismo , Animais , Córnea/fisiologia , Paquimetria Corneana , Humanos , Microscopia de Fluorescência , Técnicas de Cultura de Órgãos , Ovinos , Retalhos Cirúrgicos/fisiologia , Aderências Teciduais
18.
Ocul Surf ; 14(2): 113-20, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26804252

RESUMO

The corneal stroma contains a population of mesenchymal cells subjacent to the limbal basement membrane with characteristics of adult stem cells. These 'niche cells' support limbal epithelial stem cell viability. In culture by themselves, the niche cells display a phenotype typical of mesenchymal stem cells. These stromal stem cells exhibit a potential to differentiate to multiple cell types, including keratocytes, thus providing an abundant source of these rare cells for experimental and bioengineering applications. Stromal stem cells have also shown the ability to remodel pathological stromal tissue, suppressing inflammation and restoring transparency. Because stromal stem cells can be obtained by biopsy, they offer a potential for autologous stem cell treatment for stromal opacities. This review provides an overview of the status of work on this interesting cell population.


Assuntos
Células-Tronco , Substância Própria , Epitélio Corneano , Humanos , Limbo da Córnea
19.
Methods Mol Biol ; 1341: 285-94, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26026882

RESUMO

Human Embryonic Stem Cells (hESC) offer an important resource as a limitless supply of any differentiated cell type of the human body. Keratocytes, cells from the corneal stroma, may have the potential for restoration of vision in cell therapy and biomedical engineering applications, but these specialized cells are not readily expanded in vitro. Here we describe a two-part method to produce keratocytes from the H1 hESC cell line. The hESC cells, maintained and expanded in feeder-free culture medium are first differentiated to neural crest cells using the stromal-derived inducing activity (SDIA) of the PA6 mouse embryonic fibroblast cell line. The resulting neural crest cells are selected by their expression of cell-surface CD271 and subsequently cultured as 3D pellets in a defined differentiation medium to induce a keratocyte phenotype.


Assuntos
Técnicas de Cultura de Células/métodos , Ceratócitos da Córnea/citologia , Células-Tronco Embrionárias Humanas/citologia , Crista Neural/citologia , Animais , Diferenciação Celular , Linhagem Celular , Técnicas de Cocultura/métodos , Humanos , Camundongos
20.
Sci Rep ; 5: 16186, 2015 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-26531048

RESUMO

Human limbal epithelial cells (HLE) and corneal stromal stem cells (CSSC) reside in close proximity in vivo in the corneal limbal stem cell niche. However, HLE are typically cultured in vitro without supporting niche cells. Here, we re-create the cell-cell juxtaposition of the native environment in vitro, to provide a tool for investigation of epithelial-stromal cell interactions and to optimize HLE culture conditions for potential therapeutic application. RAFT (Real Architecture For 3D Tissue) tissue equivalents (TEs) were used as a 3-dimensional substrate for co-culturing HLE and CSSC. Our results demonstrate that a monolayer of HLE that maintained expression of p63α, ABCB5, CK8 and CK15 (HLE markers), formed on the surface of RAFT TEs within 13 days of culture. CSSC remained in close proximity to HLE and maintained expression of mesenchymal stem cell markers. This simple technique has a short preparation time of only 15 days with the onset of HLE layering and differentiation observed. Furthermore, co-cultivation of HLE with another niche cell type (CSSC) directly on RAFT TEs, eliminates the requirement for animal-derived feeder cells. RAFT TEs may be useful for future therapeutic delivery of multiple cell types to restore the limbal niche following ocular surface injury or disease.


Assuntos
Células Epiteliais/citologia , Células-Tronco Mesenquimais/citologia , 5'-Nucleotidase/metabolismo , Técnicas de Cultura de Células , Células Cultivadas , Técnicas de Cocultura , Células Epiteliais/metabolismo , Epitélio Corneano/citologia , Humanos , Imuno-Histoquímica , Queratina-15/metabolismo , Queratina-8/metabolismo , Limbo da Córnea/citologia , Células-Tronco Mesenquimais/metabolismo , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Antígenos Thy-1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...