Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Oral Dis ; 27(6): 1383-1393, 2021 Sep.
Article in English | MEDLINE | ID: mdl-32593227

ABSTRACT

BACKGROUND: Cancer stem cells are responsible for tumour progression and chemoresistance. Fibroblasts surrounding a tumour also promote progression and fibroblast "activation" is an independent prognostic marker in oral cancer. Cancer stem cells may therefore promote tumourigenesis through communication with stromal fibroblasts. METHODS: Cancer stem cells were isolated from oral cancer cell lines by adherence to fibronectin or cisplatin resistance. Fibroblasts were exposed to conditioned medium from these cells, and the activation markers, alpha smooth muscle actin and interleukin-6, were assessed using qPCR and immunofluorescence. Stem cell markers and smooth muscle actin were examined in oral cancer tissue using immunohistochemistry. RESULTS: Adherent and chemoresistant cells expressed increased levels of stem cell markers CD24, CD44 and CD29 compared with unsorted cells. Adherent cells exhibited lower growth rate, higher colony forming efficiency and increased cisplatin resistance than unsorted cells. Smooth muscle actin and Interleukin-6 expression were increased in fibroblasts exposed to conditioned medium. In oral cancer tissue, there was a positive correlation between expression of αSMA and stem cell markers. CONCLUSIONS: Adherence to fibronectin and chemoresistance isolates stem-like cells that can activate fibroblasts, which together with a correlation between markers of both in vivo, provides a mechanism by which such cells drive tumourigenesis.


Subject(s)
Carcinogenesis , Fibroblasts , Cell Line, Tumor , Cell Transformation, Neoplastic , Culture Media, Conditioned , Humans , Neoplastic Stem Cells , Stromal Cells
2.
Oral Dis ; 26(8): 1668-1676, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32562323

ABSTRACT

BACKGROUND: The expression of XCR1 receptor and its metamorphic ligand lymphotactin (hLtn) has been shown in cancers but their precise role in tumorigenesis is poorly understood including the significance of the physiologically existing hLtn monomeric (CC3) and dimeric (W55D) confirmations where the latter thought to function as the receptor antagonist. The aim of this study was to explore the functional role of bioengineered hLtn variants and the role of fibroblasts in XCR1/hLtn expression regulation in oral cancer cells (OCCL). MATERIAL AND METHODS: qRT-PCR and flow cytometry were performed to evaluate mRNA and protein expression of XCR1 and hLtn. Recombinant hLtn variants (wild-type, CC3 and W55D mutant) were designed, expressed, purified and evaluated using proliferation, adhesion and chemotaxis assays. XCR1 and hLtn expression regulation by fibroblasts was determined using indirect co-culture. XCR1 and hLtn expression in primary and metastatic OSCC tissue was assessed using immunohistochemistry. RESULTS: hLtn caused a significant decrease in OCCL XCR1 surface protein expression. hLtn CC3 mutant was highly functional facilitating proliferation and migration. Conditioned media from primary cancer-associated and senescent fibroblasts significantly upregulated XCR1 and hLtn mRNA expression in OCCL. Immunohistochemistry revealed higher XCR1 and hLtn expression in metastatic tumour deposits and surrounding stroma compared to primary OSCC tissue. CONCLUSIONS: The development of hLtn biological mutants, regulation of XCR1 expression by its ligand hLtn and crosstalk with fibroblasts are novel findings suggesting an important role for the XCR1/hLtn axis within the OSCC tumour microenvironment. These discoveries build upon previous studies and suggest that the hLtn/XCR1 axis has a significant role in stromal crosstalk and OSCC progression.


Subject(s)
Chemokines, C , Mouth Neoplasms , Cell Movement , Chemokines , Chemokines, C/genetics , Humans , Lymphokines , Mouth Neoplasms/genetics , Sialoglycoproteins , Tumor Microenvironment
3.
Eur J Oral Sci ; 126(5): 367-372, 2018 10.
Article in English | MEDLINE | ID: mdl-30070725

ABSTRACT

The periodontal pathogen Porphyromonas gingivalis can invade host cells, a virulence trait which may contribute to the persistence of infection at subgingival sites. Whilst the antibiotic protection assay has been commonly employed to investigate and quantify P. gingivalis invasion, data obtained have varied widely and a thorough investigation of the factors influencing this is lacking. We investigated the role of a number of bacterial and host-cell factors and report that the growth phase of P. gingivalis, source (laboratory strain vs. clinical strain), host-cell identity (cell line vs. primary), host-cell lysis method, and host-cell passage number had no significant effect on bacterial invasion. However, incubation time, host-cell seeding density, method of quantification (viable count vs. DNA), and whether host cells were plated or in suspension, were shown to influence invasion. Also, cells isolated by rapid adhesion to fibronectin exhibited higher levels of P. gingivalis invasion, possibly as a result of increased levels of active α5ß1 integrin. Interestingly, this may represent a population of cells with stem cell-like properties. This study provides important new information by identifying the most important factors that influence P. gingivalis invasion assays and may help to explain variations in the levels previously reported.


Subject(s)
Host-Pathogen Interactions/physiology , Porphyromonas gingivalis/growth & development , Porphyromonas gingivalis/pathogenicity , Bacterial Adhesion/drug effects , Carcinoma, Squamous Cell , Cell Line, Tumor , Cells, Cultured , DNA, Bacterial , Fibronectins/pharmacology , Humans , Periodontal Diseases/microbiology , Stem Cells
4.
J Cell Biochem ; 118(4): 878-890, 2017 04.
Article in English | MEDLINE | ID: mdl-27748539

ABSTRACT

Fibroblasts are the major cellular component of connective tissue and experience mechanical perturbations due to matrix remodelling and interstitial fluid movement. Transforming growth factor ß1 (TGF-ß1) can promote differentiation of fibroblasts in vitro to a contractile myofibroblastic phenotype characterised by the presence of α-smooth muscle actin (α-SMA) rich stress fibres. To study the role of mechanical stimulation in this process, we examined the response of primary human fibroblasts to physiological levels of fluid movement and its influence on fibroblast differentiation and responses to TGF-ß1. We reported that in both oral and dermal fibroblasts, physiological levels of fluid flow induced widespread changes in gene expression compared to static cultures, including up-regulation of genes associated with TGFß signalling and endocytosis. TGF-ß1, activin A and markers of myofibroblast differentiation including α-SMA and collagen IA1 were also increased by flow but surprisingly the combination of flow and exogenous TGF-ß1 resulted in reduced differentiation. Our findings suggest this may result from enhanced internalisation of caveolin and TGF-ß receptor II. These findings suggest that a) low levels of fluid flow induce myofibroblast differentiation and b) fluid flow antagonises the fibroblast response to pro-differentiation signals such as TGF-ß1. We propose that this may be a novel mechanism by which mechanical forces buffer responses to chemical signals in vivo, maintaining a context-specific fibroblast phenotype. J. Cell. Biochem. 118: 878-890, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Fibroblasts/cytology , Fibroblasts/physiology , Transforming Growth Factor beta1/physiology , Actins/metabolism , Active Transport, Cell Nucleus , Activins/metabolism , Caveolin 1/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Fibroblasts/drug effects , Humans , Hydrodynamics , Mouth/cytology , Mouth/metabolism , Myofibroblasts/cytology , Myofibroblasts/drug effects , Myofibroblasts/physiology , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Skin/cytology , Skin/metabolism , Transforming Growth Factor beta1/pharmacology
5.
Tissue Eng Part C Methods ; 22(12): 1108-1117, 2016 12.
Article in English | MEDLINE | ID: mdl-27846777

ABSTRACT

Tissue-engineered oral mucosal equivalents (OME) are being increasingly used to measure toxicity, drug delivery, and to model oral diseases. Current OME mainly comprise normal oral keratinocytes (NOK) cultured on top of a normal oral fibroblasts-containing matrix. However, the commercial supply of NOK is limited, restricting widespread use of these mucosal models. In addition, NOK suffer from poor longevity and donor-to-donor variability. Therefore, we constructed, characterized, and tested the functionality of OME based on commercial TERT2-immortalized oral keratinocytes (FNB6) to produce a more readily available alternative to NOK-based OME. FNB6 OME cultured at an air-to-liquid interface for 14 days exhibited expression of differentiation markers cytokeratin 13 in the suprabasal layers and cytokeratin 14 in basal layer of the epithelium. Proliferating cells were restricted to the basal epithelium, and there was immuno-positive expression of E-cadherin confirming the presence of established cell-to-cell contacts. The histology and expression of these structural markers paralleled those observed in the normal oral mucosa and NOK-based models. On stimulation with TNFα and IL-1, FNB6 OME displayed a similar global gene expression profile to NOK-based OME, with increased expression of many common pro-inflammatory molecules such as chemokines (CXCL8), cytokines (IL-6), and adhesion molecules (ICAM-1) when analyzed by gene array and quantitative PCR. Similarly, pathway analysis showed that both FNB6 and NOK models initiated similar intracellular signaling on stimulation. Gene expression in FNB6 OME was more consistent than NOK-based OME that suffered from donor variation in response to stimuli. Mucosal equivalents based on immortalized FNB6 cells are accessible, reproducible and will provide an alternative animal experimental system for studying mucosal drug delivery systems, host-pathogen interactions, and drug-induced toxicity.


Subject(s)
Biomarkers/metabolism , Fibroblasts/cytology , Keratinocytes/cytology , Mouth Mucosa/physiology , Cadherins/metabolism , Cells, Cultured , Fibroblasts/metabolism , Gene Expression Profiling , Humans , In Vitro Techniques , Keratinocytes/metabolism , Keratins/metabolism , Mouth Mucosa/cytology
6.
Infect Immun ; 84(7): 1966-1974, 2016 07.
Article in English | MEDLINE | ID: mdl-27091929

ABSTRACT

Porphyromonas gingivalis, a key periodontal pathogen, is capable of invading a variety of cells, including oral keratinocytes, by exploiting host cell receptors, including alpha-5 beta-1 (α5ß1) integrin. Previous studies have shown that P. gingivalis accelerates the cell cycle and prevents apoptosis of host cells, but it is not known whether the cell cycle phases influence bacterium-cell interactions. The cell cycle distribution of oral keratinocytes was characterized by flow cytometry and BrdU (5-bromo-2-deoxyuridine) staining following synchronization of cultures by serum starvation. The effect of cell cycle phases on P. gingivalis invasion was measured by using antibiotic protection assays and flow cytometry, and these results were correlated with gene and surface expression levels of α5 integrin and urokinase plasminogen activator receptor (uPAR). There was a positive correlation (R = 0.98) between the number of cells in S phase and P. gingivalis invasion, the organism was more highly associated with cells in S phase than with cells in G2 and G1 phases, and S-phase cells contained 10 times more bacteria than did cells that were not in S phase. Our findings also show that α5 integrin, but not uPAR, was positively correlated with cells in S phase, which is consistent with previous reports indicating that P. gingivalis invasion of cells is mediated by α5 integrin. This study shows for the first time that P. gingivalis preferentially associates with and invades cells in the S phase of the cell cycle. The mechanism of targeting stable dividing cells may have implications for the treatment of periodontal diseases and may partly explain the persistence of this organism at subgingival sites.


Subject(s)
Cell Cycle , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Host-Pathogen Interactions , Mouth Mucosa/microbiology , Porphyromonas gingivalis/physiology , Antigens, Surface/genetics , Antigens, Surface/metabolism , Biomarkers , Cells, Cultured , Endocytosis , Gene Expression , Humans , Keratinocytes/metabolism , Keratinocytes/microbiology , Periodontal Diseases/microbiology , S Phase
7.
J Oral Pathol Med ; 43(9): 667-74, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24965032

ABSTRACT

BACKGROUND: Chemokines regulate physiological and pathological leucocyte trafficking, and chemokine receptors play a role in tumorigenesis. Expression of interleukin-8 (IL-8) receptors CXCR1 and CXCR2 has been shown in oral squamous cell carcinoma (OSCC) but remains poorly characterised. This aim of this study was to investigate CXCR1 and CXCR2 expression on normal oral keratinocytes (NOKs) and oral cancer cell lines (OCCL) and their relative response when exposed to IL-8 and growth-related oncogene-α (which selectively binds CXCR2). METHODS: mRNA and protein expression was studied using RT-PCR, immunocytochemistry and flow cytometry. ELISAs were used to investigate ERK1/2 phosphorylation and MMP production, whereas a MTS-based assay was employed to study proliferation. Migration assays were carried out using modified Boyden chambers with a matrigel coating used for invasion assays. RESULTS: mRNA expression of CXCR1 and CXCR2 was seen in both NOKs and OCCL with significantly higher protein expression in OCCL. Exposure to IL-8 and GROα increased intracellular ERK phosphorylation, proliferation, migration and invasion with OCCL showing a greater response than NOKs. These effects were mediated through CXCR1 and CXCR2 (for IL-8) and CXCR2 (for GROα) as receptor-blocking antibodies significantly inhibited the responses. IL-8 and GROα also increased MMP-9 release from NOKs and OCCL with significantly higher amounts released by OCCL. However, an increase in MMP-7 production was only seen in OCCL. CONCLUSIONS: Functional CXCR1 and CXCR2 exist on normal and cancerous oral epithelial cells, and our data suggests a role for these receptors in oral cancer biology.


Subject(s)
Carcinoma, Squamous Cell/pathology , Mouth Neoplasms/pathology , Receptors, Interleukin-8A/physiology , Receptors, Interleukin-8B/physiology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Chemokine CXCL1/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Gingiva/cytology , Humans , Interleukin-8/pharmacology , Keratinocytes/cytology , Keratinocytes/drug effects , Matrix Metalloproteinase 2/drug effects , Matrix Metalloproteinase 7/drug effects , Matrix Metalloproteinase 9/drug effects , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 3/drug effects , Neoplasm Invasiveness , Phosphorylation , Receptors, Interleukin-8A/drug effects , Receptors, Interleukin-8B/drug effects
8.
Microbes Infect ; 16(4): 310-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24491281

ABSTRACT

Porphyromonas gingivalis is a Gram-negative, keystone pathogen in periodontitis that leads to tissue destruction and ultimately tooth loss. The organism is able to infect oral epithelial cells and two-dimensional (monolayer) cultures have been used to investigate this process. However, recently there has been interest in the use of three-dimensional, organotypic mucosal models to analyse infection. These models are composed of collagen-embedded fibroblasts overlain with multilayers of oral epithelial cells. In this study we report for the first time significant differences in the response of oral mucosal models to P. gingivalis infection when compared to monolayer cultures of oral epithelial cells. Intracellular survival (3-fold) and bacterial release (4-fold) of P. gingivalis was significantly increased in mucosal models compared with monolayer cultures, which may be due to the multi-layered nature and exfoliation of epithelial cells in these organotypic models. Furthermore, marked differences in the cytokine profile between infected organotypic models and monolayer cultures were observed, particularly for CXCL8 and IL6, which suggested that degradation of cytokines by P. gingivalis may be less pronounced in organotypic compared to monolayer cultures. These data suggest that use of oral mucosal models may provide a greater understanding of the host responses to P. gingivalis invasion than simple monolayer cultures.


Subject(s)
Mouth Mucosa/microbiology , Porphyromonas gingivalis/physiology , Animals , Cytokines/metabolism , Microbial Viability , Models, Theoretical , Organ Culture Techniques/methods , Porphyromonas gingivalis/growth & development , Porphyromonas gingivalis/immunology , Rats, Wistar
9.
BMC Neurosci ; 15: 1, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24380503

ABSTRACT

BACKGROUND: The neurotrophin Nerve Growth factor (NGF) is known to influence the phenotype of mature nociceptors, for example by altering synthesis of neuropeptides, and changes in NGF levels have been implicated in the pathophysiology of chronic pain conditions such as neuropathic pain. We have tested the hypothesis that after partial nerve injury, NGF accumulates within the skin and causes 'pro-nociceptive' phenotypic changes in the remaining population of sensory nerve fibres, which could underpin the development of neuropathic pain. RESULTS: Eleven days after chronic constriction injury of the rat mental nerve the intra-epidermal nerve fibre density of the chin skin from had reduced from 11.6 ± 4.9 fibres/mm to 1.0 ± 0.4 fibres/mm; this slowly recovered to 2.4 ± 2.0 fibres/mm on day 14 and 4.0 ± 0.8 fibres/mm on day 21. Cold hyperalgesia in the ipsilateral lower lip was detectable 11 days after chronic constriction injury, although at this time skin [NGF] did not differ between sides. At 14 days post-injury, there was a significantly greater [NGF] ipsilaterally compared to contralaterally (ipsilateral = 111 ± 23 pg/mg, contralateral = 69 ± 13 pg/mg), but there was no behavioural evidence of neuropathic pain at this time-point. By 21 days post-injury, skin [NGF] was elevated bilaterally and there was a significant increase in the proportion of TrkA-positive (the high-affinity NGF receptor) intra-epidermal nerve fibres that were immunolabelled for the neuropeptide Calcitonin Gene-related peptide. CONCLUSIONS: The temporal mismatch in behaviour, skin [NGF] and phenotypic changes in sensory nerve fibres indicate that increased [NGF] does not cause hyperalgesia after partial mental nerve injury, although it may contribute to the altered neurochemistry of cutaneous nerve fibres.


Subject(s)
Behavior, Animal , Nerve Fibers/pathology , Nerve Growth Factor/metabolism , Neuralgia/physiopathology , Skin/innervation , Skin/metabolism , Trigeminal Nerve Injuries/metabolism , Animals , Cell Count , Male , Nerve Fibers/metabolism , Rats , Rats, Sprague-Dawley , Skin/pathology , Trigeminal Nerve Injuries/pathology
10.
FEBS Lett ; 587(21): 3529-34, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24055471

ABSTRACT

A disintegrin and metalloprotease (ADAM) proteins are upregulated in cancer and can interact with integrin receptors. We investigated whether such interactions may have functional significance in oral squamous cell carcinoma (OSCC). ADAM 10 expression was increased in OSCC tissue and cell lines compared to normal oral mucosa. Silencing of ADAM 10 reduced migration and invasion specifically in OSCC cells over-expressing αvß6 integrin. This may result from ADAM 10-induced up-regulation of MMPs. We conclude ADAM 10 may influence OSCC invasion by functionally interacting with αvß6 integrin which we have previously shown is over expressed in OSCC.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Antigens, Neoplasm/metabolism , Carcinoma, Squamous Cell/metabolism , Integrins/metabolism , Membrane Proteins/metabolism , Mouth Mucosa/pathology , Mouth Neoplasms/metabolism , ADAM Proteins/genetics , ADAM10 Protein , Amyloid Precursor Protein Secretases/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement , Humans , Membrane Proteins/genetics , Mouth Mucosa/metabolism , Mouth Neoplasms/pathology , Up-Regulation
11.
J Med Microbiol ; 61(Pt 12): 1654-1661, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22956750

ABSTRACT

Staphylococcus aureus can invade epithelial cells, and the host-cell receptor α(5)ß(1) integrin is thought to mediate this process. The aim of this study was to investigate S. aureus invasion of epithelial cell lines derived from oral (H357), skin (UP) and nasopharyngeal (Detroit 562) sites and to determine whether any differences were due to the levels of α(5)ß(1) integrin expressed. While the adhesion and invasion of two S. aureus strains were similar in both oral and skin-derived keratinocytes, this was markedly reduced in the nasopharyngeal cell line, despite it expressing similar levels of α(5)ß(1). While this might be explainable on the basis of availability of cell receptor, adhesion to and invasion of H357 and UP cells by S. aureus were enhanced when the epithelial cells were in suspension rather than on a surface, and levels of α(5) integrin subunit mRNA were also increased. Detroit 562 cells exhibited a similar α(5) gene upregulation, but this did not result in enhanced adhesion and invasion of S. aureus. The Detroit 562 cells also showed reduced adhesion to fibronectin compared with the other cell types. This, and the low S. aureus invasion, may result from reduced α(5)ß(1) integrin activity or from variation in an as-yet-unidentified additional receptor or accessory molecule. These studies shed further light on the mechanisms of S. aureus invasion of human cells.


Subject(s)
Bacterial Adhesion , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Integrin alpha5beta1/metabolism , Staphylococcus aureus/physiology , Staphylococcus aureus/pathogenicity , Adhesins, Bacterial/metabolism , Bacterial Proteins/metabolism , Cell Line , Fibronectins/metabolism , Humans , Integrin alpha5beta1/genetics , Keratinocytes/microbiology , Mouth Mucosa/microbiology , Nasal Mucosa/microbiology , RNA, Messenger/biosynthesis , Skin/microbiology
12.
Int J Cancer ; 130(1): 40-7, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21491424

ABSTRACT

The invasion and migration of cancer cells is increasingly recognised to be influenced by factors derived from adjacent tumour-associated stroma. The contextual signals regulating stromal-tumour interactions, however, remain poorly understood. Here, we identify a role for endothelin-1 (ET-1), a mitogenic peptide elevated in a number of malignancies, in promoting pro-metastatic cross-talk between head and neck cancer cells and adjacent fibroblasts. We demonstrate that treatment of oral fibroblasts with ET-1 activates ADAM17-mediated release of epidermal growth factor receptor (EGFR) ligands, triggering EGFR signalling and increased motility in neighbouring head and neck cancer cells. ET-1-mediated paracrine transactivation of EGFR also increased cyclo-oxygenase-2 levels in the cancer cells, providing a molecular insight into the mechanisms by which the elevated levels of ET-1 observed in head and neck cancers may contribute to disease progression.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cell Communication , Cell Movement/physiology , Endothelin-1/metabolism , Epithelial Cells/pathology , Head and Neck Neoplasms/pathology , Stromal Cells/pathology , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM17 Protein , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cells, Cultured , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Endothelin-1/genetics , Epithelial Cells/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Humans , Mouth/cytology , Mouth/metabolism , Paracrine Communication , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction , Stromal Cells/metabolism
13.
J Oral Pathol Med ; 40(10): 755-61, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21615501

ABSTRACT

BACKGROUND: Integrins initiate signalling in response to the extracellular matrix (ECM), which is important in wound healing and cancer. Previous studies have shown that over-expression of the αvß6 integrin in oral squamous cell carcinoma (OSCC) cells results in enhanced motility and expression of matrix-degrading proteases, and the aim of this study was to investigate whether this is also the case for the α9ß1 integrin. METHODS: H357 OSCCcells were transfected with the α9 integrin subunit and proliferation, adhesion and migration assays were performed on these along with null vector control and wild-type cells. The effect of ligand engagement on matrix metalloproteinase expression and the plasminogen activator system was measured using ELISA and chromogenic assays. Expression of α9 integrin was examined in oral squamous cell carcinoma tissue by immunohistochemistry. RESULTS: Functionally active α9 integrin mediated specific upregulation of adhesion and migration towards the TNfn3RAA fragment of tenascin-C but reduced proliferation. Migration towards collagen I was also enhanced in transfected cells. Matrix metalloproteinase-2 and metalloproteinase-9 expression was increased upon TNfn3RAA ligand engagement. Cell surface plasmin generation was also enhanced in α9-expressing cells and was the result of enhanced expression of urokinase receptor. In normal oral mucosa, α9 integrin expression was restricted to the suprabasal and prickle cell layers, and expression was heterogeneous in tumours but present in islands infiltrating connective tissue particularly in moderately and well-differentiated lesions. CONCLUSIONS: The α9ß1 integrin may play a key role in modulation of tumour behaviour including enhanced cell migration and expression of matrix-degrading proteases.


Subject(s)
Carcinoma, Squamous Cell/pathology , Integrin alpha Chains/physiology , Integrins/physiology , Mouth Neoplasms/pathology , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation , Collagen Type I/analysis , Connective Tissue/pathology , Epithelial Cells/pathology , Extracellular Matrix/pathology , Fibrinolysin/analysis , Fibronectins/analysis , Flow Cytometry , Humans , Immunohistochemistry , Integrin alpha Chains/genetics , Integrins/genetics , Matrix Metalloproteinase 2/analysis , Matrix Metalloproteinase 9/analysis , Mouth Mucosa/pathology , Peptide Fragments/analysis , Tenascin/analysis , Transfection , Up-Regulation , Urokinase-Type Plasminogen Activator/analysis
14.
Immunology ; 132(2): 188-96, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20875079

ABSTRACT

The integrin αE(CD103)ß7 (αEß7) is expressed by intraepithelial lymphocytes, dendritic cells and regulatory T cells. It plays an important role in the mucosal immune system by retaining lymphocytes within the epithelium and is involved in graft rejection, immunity against tumours and the generation of gut-homing effector cells. In gut and breast, the ligand for αEß7 is E-cadherin but in human oral mucosa and skin, there is evidence that lymphocytes use an alternative, unknown, ligand. In the present study, the I domain of the human αE subunit, which contains the E-cadherin-binding site, was locked in a highly active, 'open' and an inactive, 'closed' conformation by the introduction of disulphide bonds and these domains were expressed as IgG Fc fusion proteins. αE fusion proteins recognize E-cadherin, the only known ligand for αEß7. This interaction was inhibited by an antibody that blocks the αE-binding site on E-cadherin and by the omission of Mn(2+) , which is essential for integrin function in vitro. The locked 'open' conformation of αE adhered to human oral and skin keratinocytes, including the E-cadherin-negative H376 cell line, and this was not inhibited by blocking antibody against the αEß7-binding site on E-cadherin, providing further evidence for the existence of an alternative ligand for αEß7 in skin and oral mucosa. The interaction with E-cadherin and the alternative ligand was Mn(2+) dependent and mediated by the metal ion-dependent coordination site (MIDAS) of the locked 'open'αE I domain, independently of the ß7 subunit.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Integrin alpha Chains/metabolism , Integrin beta Chains/metabolism , Keratinocytes/metabolism , Mouth Mucosa/cytology , Skin/cytology , Antigens, CD/chemistry , Antigens, CD/genetics , Binding Sites , Cadherins/chemistry , Cadherins/genetics , Cell Adhesion , Cell Line, Tumor , Humans , Immunity, Mucosal , Integrin alpha Chains/chemistry , Integrin alpha Chains/genetics , Integrin beta Chains/chemistry , Integrin beta Chains/genetics , Keratinocytes/cytology , Ligands , Mouth Mucosa/immunology , Mouth Mucosa/metabolism , Protein Conformation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Skin/immunology , Skin/metabolism , T-Lymphocytes/immunology
15.
FEBS Lett ; 585(1): 187-92, 2011 Jan 03.
Article in English | MEDLINE | ID: mdl-21112327

ABSTRACT

Alterations in the levels of molecules which interact with the extracellular matrix, such as integrins, are associated with invasion of oral squamous cell carcinomas (OSCC). The molecular mechanisms underlying dysregulation of integrin expression in OSCC, however, remain unclear. Here, we show that microRNA-124, a small non-coding RNA down-regulated in OSCC, is able to downregulate expression of integrin beta-1 (ITGB1) by interacting with its 3' untranslated region. Over-expression of miR-124 attenuates endogenous ITGB1 expression and reduces the adherence and motility of OSCC cells, suggesting disruption of miR-124-mediated repression of ITGB1 may be a key factor in OSCC progression.


Subject(s)
Cell Movement , Integrin beta1/genetics , MicroRNAs/genetics , 3' Untranslated Regions/genetics , Binding Sites/genetics , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Adhesion , Cell Line, Tumor , Down-Regulation , Humans , Integrin beta1/metabolism , Luciferases/genetics , Luciferases/metabolism , MicroRNAs/metabolism , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Mutagenesis, Site-Directed , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
16.
J Pathol ; 221(2): 153-63, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20225245

ABSTRACT

Chemokines are chemoattractant cytokines which act on specific receptors and play an important role in leukocyte migration as well as physiological and pathological processes. We investigated the role of the chemokine receptor XCR1 and its ligand lymphotactin (Lptn/XCL1) in the regulation of oral epithelial cell behaviour. In vitro XCR1 mRNA and cell surface protein expression was detected in normal oral keratinocytes and oral squamous cell carcinoma cell lines. Lymphotactin mediated intracellular activation of the ERK1/2 signalling pathway and stimulated migration, invasion, and proliferation of all cells through XCR1. Oral cancer cells showed a greater response to lymphotactin than normal keratinocytes and a direct relationship between receptor expression and migration, invasion, and proliferation was observed. Exposure of normal keratinocytes to lymphotactin resulted in increased adhesion to fibronectin but not collagen and stimulated MMP-2 and MMP-9 but not MMP-7 release, whereas exposure of cancer cells resulted in increased adhesion to both collagen and fibronectin and stimulated production of MMP-2, MMP-9, and MMP-7. We observed XCR1 but not lymphotactin to be expressed by epithelial cells in normal oral mucosa in vivo, whilst both were expressed and up-regulated in inflammatory oral disease and oral cancer including primary and metastatic disease. Lymphotactin mRNA and constitutive intracellular protein were detected in normal keratinocytes and oral cancer cell lines in vitro. These findings show that XCR1 and its ligand, lymphotactin, are expressed by oral epithelial cells and suggest that they play a role in regulating the behaviour of these cells.


Subject(s)
Chemokines, C/metabolism , Keratinocytes/metabolism , Mouth Neoplasms/metabolism , Receptors, G-Protein-Coupled/metabolism , Cell Line , Cell Movement , Cell Proliferation , Epithelial Cells/metabolism , Humans , Lymphocyte Activation , Mitogen-Activated Protein Kinase 3/metabolism , Mouth Neoplasms/pathology , Neoplasm Invasiveness , Phosphorylation , RNA, Messenger/metabolism
17.
J Tissue Eng Regen Med ; 3(8): 647-50, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19701934

ABSTRACT

Fibroblast-epithelium interactions are crucial for successful tissue engineering of skin and oral mucosal equivalents. In this study, we assessed early force generation in organotypic fibroblast-epithelium co-cultures, using normal human keratinocytes (NHK) and HPV16-transformed (UP) cells. During the initial 2 h period, organotypic co-cultures containing both epithelial cell types produced significantly more force than fibroblasts alone (p < 0.05). After 2 h, the epithelial contribution became diminished and did not significantly contribute to intrinsic force generation by fibroblasts, and no differences were observed when using UP vs. NHK. We then measured protease gene expression at the end of the experimental period. Distinct differences were evident in protease expression both between NHK-human skin fibroblast (HSF) vs. UP-HSF co-cultures and compared to fibroblasts alone. We conclude that whilst the very early contractile response of fibroblasts is enhanced by the overlying epithelium, this becomes diminished as the fibroblast response becomes predominant and it does contribute to tissue remodelling via regulation of protease expression.


Subject(s)
Coculture Techniques/methods , Fibroblasts/cytology , Gene Expression Regulation , Keratinocytes/cytology , Tissue Engineering/methods , Cells, Cultured , Fibroblasts/metabolism , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Peptide Hydrolases/biosynthesis , Receptors, Urokinase Plasminogen Activator/metabolism , Skin/metabolism , Stress, Mechanical , Time Factors , Urokinase-Type Plasminogen Activator/metabolism
18.
Eur J Oral Sci ; 115(6): 454-8, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18028052

ABSTRACT

Changes in the extracellular matrix, integrin expression, and protease secretion occur in wound healing and cancer and these systems are thought to play a crucial role in such processes. In this study, experiments were performed to examine the interaction of epithelial cells with the 120 kDa cell-binding fibronectin fragment. Cell migration was significantly increased in response to the 120 kDa fragment when compared with the full-length molecule, but only in cells overexpressing the beta-6 integrin (VB6). This involved the up-regulation of matrix metalloproteinases (MMPs) 2 and 9, the levels of which are increased in the supernatant from VB6 cells plated on the 120 kDa fragment. Inhibition of MMP activity with both tissue inhibitor of metalloproteinase-1 (TIMP-1) and a chemical inhibitor (Ro32-3580) resulted in a significant reduction in migration on the fibronectin fragment. In vitro this fragment can be generated by incubating full-length fibronectin with purified MMP-9. Expression of the beta6 integrin subunit, which is only seen in wound healing and cancer, enhances the motility of and MMP secretion by epithelial cells in response to the 120 kDa fibronectin fragment. These enzymes can then further degrade fibronectin to generate additional fragments, leading to the creation of a positive feedback loop, which may be of significance in disease processes.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Fibronectins/metabolism , Integrins/metabolism , Keratinocytes/cytology , Mouth Neoplasms/metabolism , Receptors, Vitronectin/metabolism , Carcinoma, Squamous Cell/enzymology , Cell Adhesion/physiology , Cell Movement/physiology , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mouth Neoplasms/enzymology
19.
Arch Dermatol Res ; 296(6): 250-7, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15503090

ABSTRACT

Staphylococcus aureus is a common wound-infecting organism which can interact with cells via the extracellular matrix protein fibronectin (FN). The aim of this study was to determine the effect of the FN-binding protein (FnBP) of S. aureus on the behaviour of a human skin keratinocytes cell line (UP). FN-coated plates and Transwell membranes were exposed to a recombinant protein encompassing the D1-D4 repeat region of S. aureus FnBPB (rFnBPBD1-D4) before cell adhesion and migration assays. The influence of this protein on migration into a wounded area, cell cycle progression and endogenous cellular FN assembly was also assessed. The rFnBPBD1-D4 protein potently inhibited S. aureus entry into UP keratinocytes but had no effect on cell adhesion to FN substrate. It inhibited UP keratinocyte Transwell migration by 17% (P= 0.04) and 31% (P=0.02) at 10 and 100 microg/ml, respectively. In the wound assay, 100 microg/ml of rFnBPBD1-D4 protein reduced the migration area by approximately 30%. No differences in cell cycle progression were observed. In the presence of rFnBPBD1-D4, most of the cellular FN matrix on the cell surface and along the cell filopodia seen in untreated cells was absent. Interaction of S. aureus FnBPs with FN may influence cell behaviour and thus play a role in delayed epithelial closure in infected healing wounds.


Subject(s)
Adhesins, Bacterial/pharmacology , Keratinocytes/physiology , Staphylococcus aureus/metabolism , Antibodies/pharmacology , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Line, Transformed , Cell Movement/drug effects , Fibronectins/metabolism , Humans , Integrin alpha5beta1/immunology , Keratinocytes/microbiology , Peptide Fragments/pharmacology , Polymers/metabolism , Recombinant Proteins/pharmacology , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology
20.
Infect Immun ; 72(10): 5668-75, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15385465

ABSTRACT

Staphylococcus aureus is among the most important human pathogens and causes various superficial and systemic infections. The ability of S. aureus to be internalized by, and survive within, host cells, such as keratinocytes, may contribute to the development of persistent or chronic infections and may finally lead to deeper tissue infections or dissemination. To examine the mechanisms of internalization of S. aureus by keratinocytes, isogenic mutants lacking fibronectin-binding proteins (FnBPs), a recombinant protein consisting of the fibronectin-binding domain of S. aureus FnBPs, and an anti-alpha5beta1 antibody were used in cocultures with immortalized keratinocytes and primary keratinocytes. We found that internalization of S. aureus by immortalized keratinocytes requires bacterial FnBPs and is mediated by the major fibronectin-binding integrin alpha5beta1. In contrast to internalization by immortalized keratinocytes, internalization of S. aureus by primary keratinocytes could occur through FnBP-dependent and -independent pathways. S. aureus clumping factor B (ClfB), which was recently determined to bind to epithelial cells, was not involved in the uptake of this bacterium by keratinocytes. The identification of an alternate uptake pathway, which is independent of S. aureus FnBPs and host cell alpha5beta1, has important implications for the design of therapies targeted to bacterial uptake by host cells.


Subject(s)
Bacterial Adhesion , Keratinocytes/cytology , Keratinocytes/microbiology , Staphylococcus aureus/physiology , Adhesins, Bacterial/metabolism , Antibodies/immunology , Antibodies/pharmacology , Antigens, Neoplasm/metabolism , Bacterial Adhesion/drug effects , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biological Transport/drug effects , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Endocytosis , Humans , Integrin alpha5beta1/antagonists & inhibitors , Integrin alpha5beta1/immunology , Integrin alpha5beta1/metabolism , Integrins/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Mutation/genetics , Skin/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...