Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 15(11): e0241569, 2020.
Article in English | MEDLINE | ID: mdl-33141875

ABSTRACT

Glaucoma filtration surgery is one of the most effective methods for lowering intraocular pressure in glaucoma. The surgery efficiently reduces intra-ocular pressure but the most common cause of failure is scarring at the incision site. This occurs in the conjunctiva/Tenon's capsule layer overlying the scleral coat of the eye. Currently used antimetabolite treatments to prevent post-surgical scarring are non-selective and are associated with potentially blinding side effects. Developing new treatments to target scarring requires both a better understanding of wound healing and scarring in the conjunctiva, and new means of delivering anti-scarring drugs locally and sustainably. By combining plastic compression of collagen gels with a soft collagen-based layer, we have developed a physiologically relevant model of the sub-epithelial bulbar conjunctiva/Tenon's capsule interface, which allows a more holistic approach to the understanding of subconjunctival tissue behaviour and local drug delivery. The biomimetic tissue hosts both primary human conjunctival fibroblasts and an immune component in the form of macrophages, morphologically and structurally mimicking the mechanical proprieties and contraction kinetics of ex vivo porcine conjunctiva. We show that our model is suitable for the screening of drugs targeting scarring and/or inflammation, and amenable to the study of local drug delivery devices that can be inserted in between the two layers of the biomimetic. We propose that this multicellular-bilayer engineered tissue will be useful to study complex biological aspects of scarring and fibrosis, including the role of inflammation, with potentially significant implications for the management of scarring following glaucoma filtration surgery and other anterior ocular segment scarring conditions. Crucially, it uniquely allows the evaluation of new means of local drug delivery within a physiologically relevant tissue mimetic, mimicking intraoperative drug delivery in vivo.


Subject(s)
Biomimetic Materials , Cicatrix/prevention & control , Conjunctiva/pathology , Postoperative Complications/prevention & control , Tenon Capsule/pathology , Animals , Biomimetics , Cell Line , Cicatrix/etiology , Cicatrix/pathology , Conjunctiva/cytology , Conjunctiva/drug effects , Conjunctiva/surgery , Drug Delivery Systems/methods , Drug Evaluation, Preclinical/methods , Feasibility Studies , Fibroblasts , Fibrosis , Filtering Surgery/adverse effects , Glaucoma/surgery , Humans , Intraoperative Care/methods , Monocytes , Postoperative Complications/etiology , Postoperative Complications/pathology , Primary Cell Culture , Swine , Tenon Capsule/drug effects , Tenon Capsule/surgery , Wound Healing/drug effects
2.
Invest Ophthalmol Vis Sci ; 56(10): 5743-50, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26325413

ABSTRACT

PURPOSE: Graves' orbitopathy (GO) is a sight-threatening autoimmune disorder causing extraocular muscle fibrosis, upper lid retraction and eye bulging due to orbital fat expansion. These clinical features are mediated by aspects of orbital fibroblasts differentiation, including adipogenesis and fibrosis. Our previous work suggested that this dual phenotype might be a manifestation of mixed cell populations, partially linked to the expression of mesenchymal stem cell (MSC) marker CD90. Thus, we set out to determine whether GO orbital fibroblasts displayed MSC properties. METHODS: Control and GO orbital fibroblasts previously characterized for CD90 and CD45 expression were analyzed by flow cytometry for classical MSC positive (CD73, CD105) and negative (CD14, CD19, HLA-DR, and CD34) markers. Graves' orbitopathy fibroblasts were tested further for their ability to undergo lineage specific differentiation following standard protocols. RESULTS: Control and GO fibroblasts strongly expressed CD73 and CD105, with a higher percentage of positive cells and stronger expression levels in GO. Neither cell type expresses CD14, CD19, and HLA-DR. Protein CD34 was expressed at low levels by 45% to 70% of the cells, with its expression significantly lower in GO cells. Graves' orbitopathy fibroblasts displayed features of osteogenesis (calcium deposits, and osteocalcin [BGLAP] and osteonectin [SPARC] expression), chondrogenesis (glycosaminoglycan production; SOX9 and aggrecan [ACAN] expression), myogenesis (α-smooth muscle actin expression), and neurogenesis (ß-III tubulin expression) upon differentiation. CONCLUSIONS: Our findings suggest that orbital fibroblasts contain a population of cells that fulfil the criteria defining MSC. This subpopulation may be increased in GO, possibly underlying the complex differentiation phenotype of the disease.


Subject(s)
Fibroblasts/metabolism , Graves Ophthalmopathy/metabolism , Mesenchymal Stem Cells/metabolism , Aggrecans/metabolism , Antigens, CD/metabolism , Biomarkers/metabolism , Case-Control Studies , Cell Differentiation/physiology , Cells, Cultured , Chondrogenesis/physiology , Fibroblasts/cytology , Graves Ophthalmopathy/pathology , HLA-DR Antigens/metabolism , Humans , Mesenchymal Stem Cells/cytology , Osteocalcin/metabolism , Osteogenesis/physiology , Osteonectin/metabolism , SOX9 Transcription Factor/metabolism
3.
PLoS One ; 9(4): e95586, 2014.
Article in English | MEDLINE | ID: mdl-24751986

ABSTRACT

Graves' orbitopathy (GO) is a disfiguring and sometimes blinding disease, characterised by inflammation and swelling of orbital tissues, with fibrosis and adipogenesis being predominant features. Little is known about the disease aetiology and the molecular mechanisms driving the phenotypic changes in orbital fibroblasts are unknown. Using fibroblasts isolated from the orbital fat of undiseased individuals or GO patients, we have established a novel in vitro model to evaluate the dual profile of GO cells in a three-dimensional collagen matrix; this pseudo-physiological 3D environment allows measurement of their contractile and adipogenic properties. GO cells contracted collagen matrices more efficiently than control cells following serum or TGFß1 stimulation, and showed a slightly increased ability to proliferate in the 3D matrix, in accordance with a fibro-proliferative phenotype. GO cells, unlike controls, also spontaneously differentiated into adipocytes in 3D cultures - confirming an intrinsic adipogenic profile. However, both control and GO cells underwent adipogenesis when cultured under pathological pressure levels. We further demonstrate that a Thy-1-low population of GO cells underlies the adipogenic - but not the contractile - phenotype and, using inhibitors, confirm that the contractile and adipogenic phenotypes are regulated by separate pathways. In view of the current lack of suitable treatment for GO, we propose that this new model testing the duality of the GO phenotype could be useful as a preclinical evaluation for the efficacy of potential treatments.


Subject(s)
Adipogenesis , Fibroblasts/pathology , Graves Ophthalmopathy/pathology , Models, Biological , Aged , Cells, Cultured , Extracellular Matrix/metabolism , Female , Humans , Male , Middle Aged , Phenotype , Pressure , Thy-1 Antigens/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...