Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Pharmacol Ther ; 242: 108349, 2023 02.
Article in English | MEDLINE | ID: mdl-36682466

ABSTRACT

Corneal diseases are one of the leading causes of moderate-to-severe visual impairment and blindness worldwide, after glaucoma, cataract, and retinal disease in overall importance. Given its tendency to affect people at a younger age than other blinding conditions such as cataract and glaucoma, corneal scarring poses a huge burden both on the individuals and society. Furthermore, corneal scarring and fibrosis disproportionately affects people in poorer and remote areas, making it a significant ophthalmic public health problem. Traditional medical strategies, such as topical corticosteroids, are not effective in preventing fibrosis or scars. Corneal transplantation, the only effective sight-restoring treatment for corneal scars, is curbed by challenges including a severe shortage of tissue, graft rejection, secondary conditions, cultural barriers, the lack of well-trained surgeons, operating rooms, and well-equipped infrastructures. Thanks to tremendous research efforts, emerging therapeutic options including gene therapy, protein therapy, cell therapy and novel molecules are in development to prevent the progression of corneal scarring and compliment the surgical options currently available for treating established corneal scars in clinics. In this article, we summarise the most relevant preclinical and clinical studies on emerging therapies for corneal scarring in recent years, showing how these approaches may prevent scarring in its early development.


Subject(s)
Cataract , Corneal Diseases , Corneal Injuries , Glaucoma , Humans , Cicatrix/therapy , Cicatrix/complications , Corneal Injuries/therapy , Corneal Injuries/complications , Corneal Diseases/drug therapy , Corneal Diseases/complications , Glaucoma/complications , Cataract/complications
2.
Int J Mol Sci ; 23(13)2022 Jun 28.
Article in English | MEDLINE | ID: mdl-35806191

ABSTRACT

Strategies that alter the pH of wounds to improve healing outcomes are an emerging area of interest. Currently, there is limited understanding of the effect of hydrogen (H+) on the functionality of skin cells during proliferation and migration, highlighting the need for research to determine the effect of pH during wound healing. This study aimed to determine the effect of acidification on the metabolic activity and migration of human immortalized keratinocytes (HaCaT) and human foreskin fibroblasts (HFF). In vitro models were used with phosphoric and citric acid buffers at a pH range between 3 and 7. Our results showed that cells were more viable in buffers with low rather than high ionic strength. A time-dependent effect of the acidification treatment was also observed with cell metabolic activity varying with treatment duration and frequency. Our results showed that a 24 h treatment and subsequent resting phase significantly improved cell proliferation and migration. This in vitro study is the first to establish a correlation between the role of acidic pH, molarity and treatment regimen in cellular activity. Our data demonstrated a positive effect of acidic pH on cell metabolic activity and migration rate, suggesting a clinical potential in indications such as wound healing.


Subject(s)
Skin , Wound Healing , Cell Movement , Cell Proliferation , Fibroblasts , Humans , Keratinocytes/metabolism , Skin/injuries
3.
Int J Mol Sci ; 22(24)2021 Dec 08.
Article in English | MEDLINE | ID: mdl-34948000

ABSTRACT

Cutaneous squamous cell carcinoma (cSCC) accounts for 25% of cutaneous malignancies diagnosed in Caucasian populations. Surgical removal in combination with radiation and chemotherapy are effective treatments for cSCC. Nevertheless, the aggressive metastatic forms of cSCC still have a relatively poor patient outcome. Studies have linked actin cytoskeletal dynamics and the Wnt/ß-catenin signaling pathway as important modulators of cSCC pathogenesis. Previous studies have also shown that the actin-remodeling protein Flightless (Flii) is a negative regulator of cSCC. The aim of this study was to investigate if the functional effects of Flii on cSCC involve the Wnt/ß-catenin signaling pathway. Flii knockdown was performed using siRNA in a human late stage aggressive metastatic cSCC cell line (MET-1) alongside analysis of Flii genetic murine models of 3-methylcholanthrene induced cSCC. Flii was increased in a MET-1 cSCC cell line and reducing Flii expression led to fewer PCNA positive cells and a concomitant reduction in cellular proliferation and symmetrical division. Knockdown of Flii led to decreased ß-catenin and a decrease in the expression of the downstream effector of ß-catenin signaling protein SOX9. 3-Methylcholanthrene (MCA)-induced cSCC in Flii overexpressing mice showed increased markers of cancer metastasis including talin and keratin-14 and a significant increase in SOX9 alongside a reduction in Flii associated protein (Flap-1). Taken together, this study demonstrates a role for Flii in regulating proteins involved in cSCC proliferation and tumor progression and suggests a potential role for Flii in aggressive metastatic cSCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Microfilament Proteins/genetics , Skin Neoplasms/genetics , Trans-Activators/genetics , Up-Regulation , Wnt Signaling Pathway , Animals , Carcinoma, Squamous Cell/chemically induced , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Methylcholanthrene/adverse effects , Mice , Skin Neoplasms/chemically induced
4.
Int J Mol Sci ; 22(15)2021 Jul 30.
Article in English | MEDLINE | ID: mdl-34361001

ABSTRACT

Epidermal progenitor cells divide symmetrically and asymmetrically to form stratified epidermis and hair follicles during late embryonic development. Flightless I (Flii), an actin remodelling protein, is implicated in Wnt/ß-cat and integrin signalling pathways that govern cell division. This study investigated the effect of altering Flii on the divisional orientation of epidermal progenitor cells (EpSCs) in the basal layer during late murine embryonic development and early adolescence. The effect of altering Flii expression on asymmetric vs. symmetric division was assessed in vitro in adult human primary keratinocytes and in vivo at late embryonic development stages (E16, E17 and E19) as well as adolescence (P21 day-old) in mice with altered Flii expression (Flii knockdown: Flii+/-, wild type: WT, transgenic Flii overexpressing: FliiTg/Tg) using Western blot and immunohistochemistry. Flii+/- embryonic skin showed increased asymmetrical cell division of EpSCs with an increase in epidermal stratification and elevated talin, activated-Itgb1 and Par3 expression. FliiTg/Tg led to increased symmetrical cell division of EpSCs with increased cell proliferation rate, an elevated epidermal SOX9, Flap1 and ß-cat expression, a thinner epidermis, but increased hair follicle number and depth. Flii promotes symmetric division of epidermal progenitor cells during murine embryonic development.


Subject(s)
Cell Division , Microfilament Proteins/genetics , Mouse Embryonic Stem Cells/metabolism , Skin/metabolism , Trans-Activators/genetics , Animals , Cells, Cultured , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Mice, Inbred BALB C , Microfilament Proteins/metabolism , Mouse Embryonic Stem Cells/cytology , SOX9 Transcription Factor/metabolism , Skin/embryology , Trans-Activators/metabolism , beta Catenin/metabolism
5.
Adv Wound Care (New Rochelle) ; 9(4): 161-173, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32117580

ABSTRACT

Objective: Activation of epidermal stem cells (EpSCs) from their quiescent niche is an integral component of wound reepithelialization and involves Wnt/ß-catenin (ß-Cat) signaling and remodeling of the actin cytoskeleton. The aim of this study was to investigate the effect of Flightless I (Flii), a cytoskeletal protein and inhibitor of wound healing, on EpSC activation during wound repair. Approach: Genetically modified Flii mice (Flii knockdown: Flii+/- , wild type: WT, Flii overexpressing: FliiTg/Tg ) received two incisional wounds along the lateral axis of the dorsal skin. Indicators of EpSC activation (epidermal growth factor receptor 1 [EGFR1], leucine-rich repeats and immunoglobulin-like domains-1 [Lrig1], K14), Wnt/ß-Cat signaling (Lgr6, Flap2, ß-Cat, and axis inhibition protein 2 [Axin2]), and cell proliferation (proliferating cell nuclear antigen [PCNA]) were assessed using immunohistochemistry. ß-Cat stabilization was examined using western blotting with cell cycling and differentiation of isolated CD34+ITGA6high EpSCs examined using real time-quantitative polymerase chain reaction after treatment with wound-conditioned media. Results:Flii+/- led to increased numbers of activated EpSCs expressing PCNA, elevated EGFR1, and decreased Lrig1. EpSCs in Flii+/- hair follicle niches adjacent to the wounds also showed expression of Wnt-activation markers including increased ß-Cat and Lgr6, and decreased Axin2. EpSCs (CD34+ITGA6high) isolated from Flii+/- unwounded skin showed elevated expression of cell-cycling genes including ΔNp63, filaggrin (Fila), involucrin (Invo), cyclin D1 (Ccnd1), and cell-division cycle protein-20 (Cdc20); and elevated ΔNp63 and Invo after treatment with wound-conditioned media compared with WT and FliiTg/Tg counterparts. Innovation: Flii was identified as an inhibitor of EpSC activation that may explain its negative effects on wound reepithelialization. Conclusion: Flii may inhibit EpSC activation by interrupting Wnt/ß-Cat signaling. Strategies that reduce Flii may increase activation of EpSCs and promote reepithelialization of wounds.


Subject(s)
Epidermal Cells/metabolism , Microfilament Proteins/metabolism , Stem Cell Niche/genetics , Stem Cells/metabolism , Trans-Activators/metabolism , Wound Healing/genetics , Animals , Cell Proliferation/genetics , Disease Models, Animal , Female , Gene Knockdown Techniques , Mice , Mice, Inbred BALB C , Mice, Transgenic , Microfilament Proteins/genetics , Proliferating Cell Nuclear Antigen/metabolism , Skin/injuries , Skin/metabolism , Trans-Activators/genetics , Wnt Signaling Pathway/genetics , beta Catenin/metabolism
6.
Front Immunol ; 9: 1833, 2018.
Article in English | MEDLINE | ID: mdl-30147695

ABSTRACT

Atopic dermatitis (AD) is a chronic pruritic inflammatory skin disease characterized by excessive inflammation and disrupted skin barrier function. Although the etiology of AD is not completely understood, clinical and basic studies suggest increasing involvement of autoantibodies against intracellular proteins. An actin remodeling protein, Flightless I (Flii), has been shown to promote development of inflammatory mediated skin conditions and impairment of skin barrier development and function. Here, we sought to determine the effect of altering Flii expression on the development of AD and its contribution to autoimmune aspects of inflammatory skin conditions. Ovalbumin (OVA)-induced AD skin-like disease was induced in Flii heterozygous (Flii+/- ), wild-type (Flii+/+ ), and Flii transgenic (FliiTg/Tg ) mice by epicutaneous exposure to OVA for 3 weeks; each week was separated by 2-week resting period. Reduced Flii expression resulted in decreased disease severity and tissue inflammation as determined by histology, lymphocytic, and mast cell infiltrate and increased anti-inflammatory IL-10 cytokine levels and a marked IFN-γ Th1 response. In contrast, Flii over-expression lead to a Th2 skewed response characterized by increased pro-inflammatory TNF-α cytokine production, Th2 chemokine levels, and Th2 cell numbers. Sera from OVA-induced AD skin-like disease Flii+/- mice showed a decreased level of autoreactivity while sera from FliiTg/Tg mice counterparts showed an altered autoantibody profile with strong nuclear localization favoring development of a more severe disease. These findings demonstrate autoimmune responses in this model of OVA-induced AD-like skin disease and suggest that Flii is a novel target, whose manipulation could be a potential approach for the treatment of patients with AD.


Subject(s)
Dermatitis, Atopic/immunology , Inflammation/immunology , Microfilament Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Skin/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Allergens/immunology , Animals , Autoantibodies/blood , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Microfilament Proteins/genetics , Ovalbumin/immunology , Receptors, Cytoplasmic and Nuclear/genetics , Trans-Activators
7.
Int J Mol Sci ; 19(7)2018 07 10.
Article in English | MEDLINE | ID: mdl-29996558

ABSTRACT

Wound healing is an increasing clinical problem involving substantial morbidity, mortality, and rising health care costs. Leucine-rich repeat flightless-interacting protein-1 (LRRFIP-1) regulates toll-like receptor (TLR)-mediated inflammation, suggesting a potential role in the healing of wounds. We sought to determine the role of LRRFIP-1 in wound repair and whether the exogenous addition of recombinant LRRFIP-1 (rLRRFIP-1) affected healing responses. Using a model of full-thickness incisional acute wounds in BALB/c mice, we investigated the effect of wounding on LRRFIP-1 expression. The effect of rLRRFIP-1 on cellular proliferation, inflammation, and collagen deposition was also investigated. LRRFIP-1 was upregulated in response to wounding, was found to directly associate with flightless I (Flii), and significantly increased cellular proliferation both in vitro and in vivo. rLRRFIP-1 reduced Flii expression in wounds in vivo and resulted in significantly improved healing with a concurrent dampening of TLR4-mediated inflammation and improved collagen deposition. Additionally, decreased levels of TGF-ß1 and increased levels of TGF-ß3 were observed in rLRRFIP-1-treated wounds suggesting a possible antiscarring effect of rLRRFIP-1. Further studies are required to elucidate if the mechanisms behind LRRFIP-1 action in wound repair are independent of Flii. However, these results identify rLRRFIP-1 as a possible treatment modality for improved healing of acute wounds.


Subject(s)
Inflammation/metabolism , Microfilament Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Skin/injuries , Wound Healing , Animals , Cell Line , Cell Proliferation , Collagen/metabolism , Disease Models, Animal , Female , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Mice, Inbred BALB C , Recombinant Proteins/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptors/metabolism , Trans-Activators , Up-Regulation
8.
Stem Cells Dev ; 25(10): 749-59, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27021878

ABSTRACT

Cutaneous stem cells (CSCs) orchestrate the homeostasis and regeneration of mammalian skin. Epithelial CSCs have been isolated and characterized from the skin and hold great potential for tissue engineering and clinical applications. The actin cytoskeleton is known to regulate cell adhesion and motility through its intricate participation in signal transduction and structural modifications. The dynamics of actin cytoskeleton can directly influence CSCs behaviors including tissue morphogenesis, homeostasis, niche maintenance, activation, and wound repair. Various regulators of the actin cytoskeleton including kinases, actin-remodeling proteins, paracrine signals, and micro-RNAs collaborate and contribute to epithelial CSC proliferation, adhesion, and differentiation. This review brings together the latest mechanistic insights into how the actin cytoskeleton participates in the regulation of epithelial CSCs during development, homeostasis, and wound repair.


Subject(s)
Actin Cytoskeleton/metabolism , Epithelial Cells/cytology , Skin/cytology , Stem Cells/cytology , Animals , Humans , Stem Cell Transplantation
9.
Oncotarget ; 6(34): 36426-40, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26497552

ABSTRACT

Flightless I (Flii) is an actin remodeling protein that affects cellular processes including adhesion, proliferation and migration. In order to determine the role of Flii during carcinogenesis, squamous cell carcinomas (SCCs) were induced in Flii heterozygous (Flii+/-), wild-type and Flii overexpressing (FliiTg/Tg) mice by intradermal injection of 3-methylcholanthrene (MCA). Flii levels were further assessed in biopsies from human SCCs and the human SCC cell line (MET-1) was used to determine the effect of Flii on cellular invasion. Flii was highly expressed in human SCC biopsies particularly by the invading cells at the tumor edge. FliiTg/Tg mice developed large, aggressive SCCs in response to MCA. In contrast Flii+/- mice had significantly smaller tumors that were less invasive. Intradermal injection of Flii neutralizing antibodies during SCC initiation and progression significantly reduced the size of the tumors and, in vitro, decreased cellular sphere formation and invasion. Analysis of the tumors from the Flii overexpressing mice showed reduced caspase I and annexin V expression suggesting Flii may negatively regulate apoptosis within these tumors. These studies therefore suggest that Flii enhances SCC tumor progression by decreasing apoptosis and enhancing tumor cell invasion. Targeting Flii may be a potential strategy for reducing the severity of SCCs.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cytoskeletal Proteins/metabolism , Microfilament Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Animals , Apoptosis/physiology , Carcinoma, Squamous Cell/genetics , Carrier Proteins , Cytoskeletal Proteins/genetics , Disease Progression , Female , Heterografts , Humans , Mice , Mice, Inbred BALB C , Microfilament Proteins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Skin Neoplasms/genetics , Trans-Activators
10.
J Pathol ; 232(5): 541-52, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24375017

ABSTRACT

Development of an intact epidermis is critical for maintaining the integrity of the skin. Patients with epidermolysis bullosa (EB) experience multiple erosions, which breach the epidermal barrier and lead to increased microbial colocalization of wounds, infections and sepsis. The cytoskeletal protein Flightless I (Flii) is a known regulator of both development and wound healing. Using Flii(+/-), WT and Flii(Tg/Tg) mice, we investigated the effect of altering Flii levels in embryos and adult mice on the development of the epidermal barrier and, consequently, how this affects the integrity of the skin in EB. Flii over-expression resulted in delayed formation of the epidermal barrier in embryos and decreased expression of tight junction (TJ) proteins Claudin-1 and ZO-2. Increased intercellular space and transepidermal water loss was observed in Flii(Tg)(/Tg) adult mouse skin, while Flii(Tg/Tg) keratinocytes showed altered TJ protein localization and reduced transepithelial resistance. Flii is increased in the blistered skin of patients with EB, and over-expression of Flii in experimental EBA showed impaired Claudin-1 and -4 TJ protein expression and delayed recovery of functional barrier post-blistering. Immunoprecipitation confirmed Flii associated with TJ proteins and in vivo actin assays showed that the effect of Flii on actin polymerization underpinned the impaired barrier function observed in Flii(Tg/Tg) mice. These results therefore demonstrate an important role for Flii in the development and regulation of the epidermal barrier, which may contribute to the impaired healing and skin fragility of EB patients.


Subject(s)
Blister/metabolism , Epidermis/metabolism , Epidermolysis Bullosa/metabolism , Proto-Oncogene Protein c-fli-1/deficiency , Proto-Oncogene Protein c-fli-1/metabolism , Wound Healing , Actins/metabolism , Animals , Blister/genetics , Blister/pathology , Cells, Cultured , Disease Models, Animal , Electric Impedance , Epidermis/pathology , Epidermolysis Bullosa/genetics , Epidermolysis Bullosa/pathology , Genotype , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Permeability , Phenotype , Polymerization , Proto-Oncogene Protein c-fli-1/genetics , Tight Junction Proteins/metabolism , Tight Junctions/metabolism , Tight Junctions/pathology , Time Factors , Up-Regulation , Water Loss, Insensible
SELECTION OF CITATIONS
SEARCH DETAIL
...