Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Hyaluronan Fragments Improve Wound Healing on In Vitro Cutaneous Model through P2X7 Purinoreceptor Basal Activation: Role of Molecular Weight

  • Kamelia Ghazi,

    Affiliation Chimie-Toxicologie Analytique et Cellulaire (EA 4463), Université Paris Descartes, Sorbonne Paris Cité, Faculté de Pharmacie, Paris, France

  • Uriell Deng-Pichon,

    Affiliation Chimie-Toxicologie Analytique et Cellulaire (EA 4463), Université Paris Descartes, Sorbonne Paris Cité, Faculté de Pharmacie, Paris, France

  • Jean-Michel Warnet,

    Affiliation Chimie-Toxicologie Analytique et Cellulaire (EA 4463), Université Paris Descartes, Sorbonne Paris Cité, Faculté de Pharmacie, Paris, France

  • Patrice Rat

    patrice.rat@parisdescartes.fr

    Affiliation Chimie-Toxicologie Analytique et Cellulaire (EA 4463), Université Paris Descartes, Sorbonne Paris Cité, Faculté de Pharmacie, Paris, France

Abstract

Background

hyaluronan biopolymer is used in dermatology but the underlying mechanism and the impact of its molecular weight have not yet been investigated in skin wound healing. The aim of our work was to study the role of HA molecular weight in the proliferative phase of wound healing and to understand how this physiological biopolymer acts to promote wound healing on a human keratinocyte in vitro model.

Methodology and Findings

wound healing closure was evaluated using scratch test assay, cell proliferation by counting cell with haemocytometer, expression of CD44 and ZO-1 (protein present in tight junctions specific of epithelia) using flow cytometry, and P2X7 receptor activation on living using a cytoflurometric method. Our study showed that medium hyaluronan fragment (MMW-HA, between 100 and 300 kDa) induced a significant increase in wound closure, increased ZO-1 protein expression and induced a slight activation of P2X7 receptor, contrary to high (between 1000 and 1400 kDa) and low (between 5 and 20 kDa) molecular hyaluronan fragments that had no healing effects. Basal activation of P2X7 receptor is already known to stimulate cell proliferation and this activation in our model plays a pivotal role in MMW-HA-induced wound healing. Indeed, we showed that use of BBG, a specific inhibitor of P2X7 receptor, blocked completely the beneficial effects of MMW-HA on wound healing.

Conclusion

taken together, our results showed for the first time the relationship between P2X7 receptor and hyaluronan in wound healing, and that topical use of MMW-HA (fragment between 100 and 300 kDa) could represent a new therapeutic strategy to promote healing.

Introduction

Hyaluronan (HA), the major component of the ECM, is the only linear non-sulfated glycosaminoglycane composed of alternating β-1,4-glucuronic acid and β-1,3-N-acetylglucosamin [1]; [2]. HA molecular weight varies from 105 to 107 Da before being progressively degraded into smaller fragments in the ECM [3]. HA was first isolated from the vitreous humour of bovine eyes [4]. It is also present in synovial fluid [5] and is distributed ubiquitously in vertebrate tissues. The major physiological roles of high molecular weight HA in human body are to lubricate articulations and to maintain the cohesion and structure of epithelium. HA performs many pivotal structural and physiological functions in skin repair following injury [3]; [6]. Wound healing consists of 3 processes: inflammation, proliferation and remodelling. During the inflammatory phase of wound healing, hyaluronan accumulates in the wound bed. Its major function is the modulation of inflammatory cell and dermal fibroblast activities, e.g. cellular migration, proinflammatory cytokine synthesis and the phagocytosis of invading microbes [6]. Low molecular weight HA binds to TLR4 to induce inflammatory responses stimulating IL-6, TNFα and IL-1β [7]. Proliferative step is a set of complex biological responses requiring extracellular matrix (ECM) and cytoskeletal remodelling, signal cascades, and gene regulation to induce fibroblast and keratinocyte migration and proliferation. The levels of HA synthesized by both fibroblasts and keratinocytes are elevated during re-epithelialization. HA binds to cell via several receptors: CD44, RHAMM and ICAM. CD44 is recognized as the major receptor of HA [8]; [9]. The interaction of HA with CD44 induces many physiological events, such as cell migration and proliferation [10]; [11]. CD44 is localised in lipid raft domains which are plasma membrane domains that contain high levels of cholesterol and sphingolipids. Proteins such as receptors involved in cell signalling are enriched in lipid rafts [12]. P2X7 receptor, contained in lipid rafts, is a purinergic receptor; it was first cloned from rat brain [13] and, subsequently, has been found to be expressed in microglia, neurons, and astrocytes [14][16]. P2X7 receptor was activated by extracellular ATP and its analogues [17]; [18] and blocked specifically by Brillant Blue G (BBG) [19][21]. Our laboratory showed that HA of molecular weight superior to 100 kDa modulates the activation of P2X7 receptor via binding to CD44 in ophthalmic cells [22]; [23]. P2X7 receptor has been reported to play a role in cell-to-cell contact [19] and cell proliferation [24]. Moreover P2X7 receptor activation seems to be associated to corneal wound healing [25], Mankus et al showed that P2X7 receptor activation enhanced cell migration [26].

The aim of our work was to study the role of HA molecular weight in the proliferative phase of wound healing and understand how this physiological biopolymer acts to promote wound healing on a human keratinocyte in vitro model. We evaluated wound healing closure, cell proliferation, expression of CD44 and ZO-1 (protein present in tight junctions specific of epithelia), and P2X7 receptor activation after incubation of cells with a native HA and two different fragments of HA.

thumbnail
Figure 1. Scratch wound assay.

Monolayer was wounded by manual scratch with a pipette tip. Solutions containing different HA at 0.2% in culture medium with 2.5% of FCS were distributed at Day 0 and cultures were kept at 37°C for 24 hours (Day1). A: control, B: HMW-HA, C: MMW-HA, D: LMW-HA. Histogram above represents width of the wound measured for each wound at D0 and D1. Results are expressed as a ratio of D1 on D0. Wound healing area: Results are expressed as a ratio of D1 on D0 after image analysis; % of wound area represents the ration of wound area at D1/wound area at D0. We show that only MMW-HA induced a significant wound healing compared to control and other HA (***: p<0.001 compared to culture medium, n = 3). The wound healing factor is three times higher compared to control.

https://doi.org/10.1371/journal.pone.0048351.g001

Results

Scratch Wound Assay

To study wound healing, we use three different HA molecular weight (table 1). Results showed that, only MMW-HA induced a statistically significant effect on wound healing: the wound healing factor is three times higher compared to control (Figure 1). HMW-HA and LMW-HA had no effect on wound healing compared to control. % of wound area was 77.70%, 59.24%, 5.83% and 43.58% respectively for culture medium, HMW-HA, MMW-HA and LMW-HA.

thumbnail
Figure 2. Cell proliferation.

Cells were cultured in medium with 2.5% FCS containing HA HMW-HA or MMW-HA or LMW-HA at 0.2%. Proliferation was analyzed by counting cells during 6 days. circle: cell culture medium, triangle: HMW-HA, square: MMW-HA, lozenge: LMW-HA. Our results show that from day 5 only MMW-HA induced a significative increase on cell proliferation compared to control, HMW-HA and LMW-HA have no effect on cell proliferation (***: p<0.001 compared to culture medium, n = 3).

https://doi.org/10.1371/journal.pone.0048351.g002

thumbnail
Figure 3. ZO-1 protein expression.

ZO-1 expression was evaluated using flow cytometry after incubation with HMW-HA or MMW-HA or LMW-HA at 0.2% for 24 hours. Results showed a strong increase in ZO-1 expression after MMW-HA incubation. A: cell culture medium, B: HMW-HA, C: MMW-HA, D: LMW-HA (n = 3). Data show that MMW-HA increase the expression of ZO-1 protein compared to control.

https://doi.org/10.1371/journal.pone.0048351.g003

Cell Proliferation

The results show that from Day 5, MMW-HA increased significantly cell proliferation compared to control. High and low molecular weight hyaluronan had no significant effect on cell proliferation (Figure 2).

thumbnail
Figure 4. CD44 protein expression.

CD44 expression was evaluated using flow cytometry after incubation with HMW-HA or MMW-HA or LMW-HA at 0.2% for 24 hours. Results showed a very slight effect on CD44 expression level after MMW-HA incubation. A:control, B: HMW-HA, C: MMW-HA, D: LMW-HA (n = 3). Results show that CD44 expression is not modified with HA regardless the molecular weight.

https://doi.org/10.1371/journal.pone.0048351.g004

Tight Junction: ZO-1 Expression

As shown in figure 3, ZO-1 expression was strongly increased when the cells were incubated with MMW-HA (mean fluorescence = 36418.6) compared to control (mean fluorescence, = 17951.3). HMW-HA and LMW-HA had no effect on ZO-1 expression compared to control.

thumbnail
Figure 5. P2X7 receptor activation.

Cells were pre-incubated or not with a specific inhibitor of P2X7 (BBG) at 20 µM for 20 minutes before incubation with HMW-HA or MMW-HA or LMW-HA at 0.2% for 24-hours. P2X7 activation was evaluated by YO-PRO-1 dye uptake. A: cell culture medium, B: positive control, C: HMW-HA without specific inhibitor of P2X7 (BBG), D: HMW-HA with specific inhibitor of P2X7 (BBG), E: MMW-HA without specific inhibitor of P2X7 (BBG), F: MMW-HA with specific inhibitor of P2X7 (BBG), G: FMW-HA without specific inhibitor of P2X7 (BBG), H: LMW-HA with specific inhibitor of P2X7 (BBG). Incubation with MMW-HA induces slight increase in P2X7 activation compared to control and other HA (n = 18). Moreover preincubation of cell with specific inhibitor of P2X7 (BBG) inhibits MMW-HA effect on P2X7 receptor.

https://doi.org/10.1371/journal.pone.0048351.g005

thumbnail
Figure 6. Cell viability.

Cells were preincubated or not with specific inhibitor of P2X7 (BBG) at 20 µM for 20 minutes before incubation with MMW-HA at 0.2% for 24-hours. Cell viability was evaluated by neutral red test after incubation with MMW-HA. Grey: without specific inhibitor of P2X7 (BBG), black: with specific inhibitor of P2X7 (BBG). Slight activation of P2X7 we observe with MMW-HA has no effect on cell viability (n = 18).

https://doi.org/10.1371/journal.pone.0048351.g006

Hyaluronan Receptor: CD44 Expression

As shown in figure 4, the three HA had no effect on CD44 expression compared to control.

thumbnail
Figure 7. Scratch wound assay.

Monolayer was wounded by manual scratch with a pipette tip and cells were preincubated with BBG at 20 µM for 20minutes before HA incubation. Different solutions containing different HA at 0.2% in culture medium with 2.5% of FCS are distributed (Day0) and culture was kept at 37°C for 24 hours (Day1). A: control without specific inhibitor of P2X7 (BBG), B: MMW-HA with without specific inhibitor of P2X7 (BBG), C: MMW-HA with without specific inhibitor of P2X7 (BBG). % of wound area represents the ration of wound area at D1/wound area at D0. We show that cell preincubation with BBG blocks beneficial effect of MMH-HA on wound healing (***: p<0.001 compared to culture medium, n = 3).

https://doi.org/10.1371/journal.pone.0048351.g007

P2X7 Receptor Activation

In cells where P2X7 receptor is activated, we can observe a green fluorescent signal. As shown in figure 5, incubation of cells with MMW-HA induced a slight activation of P2X7 receptor compared to control and this activation was inhibited by BBG, a specific inhibitor of P2X7. HMW-HA and LMW-HA had no effect on P2X7 receptor activation compared to control.

Cell Viability

HMW-HA, MMW-HA and LMW-HA didn’t induce any decrease in cell viability (figure 6).

P2X7 Activation and Wound Healing

To know if P2X7 receptor plays a role in wound healing induced by MMW-HA, cells were incubated with BBG at 20 µM for 20 minutes before MMW-HA incubation. Results showed that preincubation with specific inhibitor of P2X7 (BBG) blocked the beneficial effects of MMW-HA on wound healing (figure 7). % of wound area was 80.03%, 37.08% and 67.38% respectively for culture medium, MMW-HA without BBG and MMW-HA with BBG.

Discussion

In clinics, some sticking plasters are composed of HA for its hygroscopic properties (hydrated HA can contain up to 1000-fold more water than its own weight). High molecular weight HA has the ability to absorb water and then it keeps the wound in a moist environment favourable to healing. Nevertheless, HA has also biological properties that could explain its role in wound healing. We found that from a physiobiological point of view, MMW-HA induced a much better healing rate than LMW-HA and HMW-HA. Indeed, we demonstrate that native HA, in spite of its essential hygroscopic role, is not the best HA to promote wound healing, and smaller molecular weight (300 kDa) HA seem to play an important biological role in wound healing. Studies have shown that HA modulate cell proliferation [10]; [27], depending on the molecular weight. Hyaluronidases, that are responsible for the degradation of hyaluronic acid, stimulate cell proliferation [28] leading to the conclusion that high molecular weight HA must be degraded to induce cell proliferation. The importance of HA degradation for cell proliferation was confirmed by a recent study showing that high molecular weight HA decreases astrocytes proliferation, but its degradation into smaller fragments induces astrocytes proliferation and activation [29]. We observed that HMW-HA didn’t induce cell proliferation, showing that hyaluronidases may not be present in our cell model. On the contrary, MMW-HA induced cell proliferation compared to control. LMW-HA had no effect on cell proliferation but this was not surprising since low molecular weight HA is pro-inflammatory [7]; [30]. So, the use of this HA could potentiate inflammation in wound leading to tissue damage. Considering these data, we focused our attention on MMW-HA.

Cell-cell junction/adhesion is an important factor for wound healing. Tight junctions play a central role in close cell-cell adhesion in simple epithelia and endothelia, connecting neighbouring cells in a controlled manner. It is composed of transmembrane proteins including claudin family proteins, occludin like proteins, junctional adhesion molecules (JAMs) and plaque proteins such as ZO-1 [31]. Besides, it plays a crucial role in the formation of the epidermal diffusion barrier. The expression of the tight junction proteins ZO-1 has recently been studied in a human skin organ culture model [32], showing ZO-1 expression early during wound healing. In our model, incubation of cells with MMW-HA induced an increase in ZO-1 expression. Not only MMW-HA stimulated cell proliferation and wound healing, but it also increased the expression of tight junctions, showing that the rebuilt epithelium monolayer possessed functional properties.

To understand how MMW-HA promotes wound closure and cell proliferation, we studied CD44 expression and P2X7 receptor activation. The results we obtained show that incubation of cells with MMW-HA has no significative effect on CD44 receptor expression (major receptor of HA). This indicates that the healing we observed with MMW-HA is not linked to the modulation of CD44 receptor expression, but probably due to other cell signalling pathway. Recently, several studies have highlighted the possibility that HA binding to CD44 induces its redistribution into lipid rafts, with multiple signal molecules being recruited and assembled to facilitate the transduction of signals [33]; [34]. Among the receptors present in lipid raft, there is the P2X7 purinergic receptor. First, P2X7 receptor was identified as a cytolytic receptor [13], but it was later shown that basal activation of P2X7 increased mitochondrial calcium and promoted cell proliferation [35]; [36]. P2X7 receptor activation has a dual role: cell proliferation after basal activation and cytolysis after high activation. The role of P2X7 receptor activation in wound healing was first highlighted in cornea [25]. The same authors showed that in P2X7−/− mice, there was a downward trend in the rate of epithelial wound repair [37]. Our results show that MMW-HA induced a slight activation of P2X7, and this slight activation was not accompanied with a loss of cell viability. This basal P2X7 activation plays a pivotal role in MMW-HA-induced wound healing since BBG, a specific inhibitor of P2X7 receptor activation, inhibited wound closure promoted by MMW-HA. These results showed that when P2X7 receptor is blocked, MMW-HA lost its healing properties. According to our results, MMW-HA (166 kDa) seems to be the best healing agent compared to LMW-HA and HLW-HA. Besides, HA with molecular weight inferior to 200 kDa has been shown to stimulate β-defensin-2 on human skin cells [38]. β-defensin-2, a peptide produced by different epithelial cells, exerts a strong antimicrobial activity against Gram-negative bacteria and Candida albicans, together with a good bacteriostatic activity against Gram-positive bacteria [39]. MMW-HA combines proliferative stimulating effects and indirect antimicrobial activity. These both properties confer to MMW-HA an interesting role in wound healing.

Our study allowed us to better understand the mechanism by which hyaluronan acts in wound healing (see above summary diagram, figure 8).

thumbnail
Figure 8. Summary diagram.

MMW-HA induces wound healing via P2X7 basal activation.

https://doi.org/10.1371/journal.pone.0048351.g008

We demonstrated that native HA is not the best biopolymer to stimulate wound healing and cell proliferation, but rather HA at 166 kDa represents an interesting therapeutic strategy as it promotes in vitro the rebuilt of a functional epithelium monolayer. We highlighted for the first time the relationship between HA at 166 kDa and P2X7 receptor basal activation in wound healing. Together, these results show that topical use of medium molecular weight hyaluronan in sticking plasters should be taken into consideration to promote tissue repair and wound healing.

Materials and Methods

Reagents

Chemicals, cell culture reagents and fluorescent dyes were purchased from Sigma-Aldrich (Saint-Quentin-Fallavier, France), Eurobio (Les Ulis, France) and Invitrogen (Cergy Pontoise, France), respectively. Different HA were obtained by biotechnology processes (Laboratoire Soliance, Pomacle, France): one native HA (HMW-HA) around 1 MDa, and two smaller fragments, one medium molecular weight HA (MMW-HA) around 150 kDa and one low moleculear weight HA (LMW-HA) around 15 kDa. Molecular weights were calculated according to European Pharmacopoeia guidelines 6.3. Hyaluronan was used at 0.2%; this concentration was tested according our previous study [23] which are showed that hyaluronan at this concentration had an effect on corneal cells.

Experimental Procedures

Cell culture.

HaCat cells (human skin keratinocytes, [40]) were cultured under standard conditions (moist atmosphere of 5% CO2 at 37°C) in Dulbecco’s minimum essential medium (DMEM) supplemented with 10% foetal calf serum (FCS), 2 mM L-glutamine, 50 IU/ml penicillin, and 50 IU/ml streptomycin. The medium was changed every three days. Confluent cultures were removed by trypsin incubation, and then cells were counted. They were seeded in culture microplates at a density of 250,000 cells per mL.

Cells were pre-incubated or not with a specific inhibitor of P2X7 (brilliant Blue G- BBG) at 20 µM for 20 minutes before incubation with HMW-HA or MMW-HA or LMW-HA at 0.2% for 24-hours.

Scratch wound assay.

According to Buonomo’s method [41][43], the cells were seeded into 6-well culture microplates, and culture was kept at 37°C for 72 hours. The medium culture was removed and the cells were rinsed with phosphate buffer saline (PBS) and incubated with culture medium without FCS during 24 hours. After 24 hours, medium was removed and the cells were washed and wounded by manual scratch with a pipette tip. The cells were rinsed with PBS and pictures of the wounds were taken using Nikon Coolpix camera. Solutions containing different HA at 0.2% in culture medium with 2.5% of FCS were distributed. Cultures were kept at 37°C for 24 hours. At D1 (+24h), medium was removed and the cells were washed before taking pictures of each wound. % of wound area was measured using Aphelion Dev image processing and analysis software developed by ADCIS S.A.

Cell proliferation.

Cells were seeded with different HA at 0.2% in culture medium at 80,000 cells per mL into 48-well culture microplates. Each day, the cells were removed with trypsin and counted for 6 days with haemocytometer.

Tight junction: ZO-1 expression.

ZO-1 expression was evaluated using flow cytometry [44]. Cells were seeded into 6-well culture microplates, culture were kept at 37°C for 24 hours. The cells were incubated with different HA at 0.2% in medium with 2.5% of FCS for 24 hours. After 24 hours, medium was removed and the cells were washed and removed by trypsin. The cells were fixed with 1% paraformaldehyde in PBS. The cells were washed with PBS, and incubated for 30 minutes at 4°C with ZO-1 polyclonal antibodies (ZYMED Laboratories) diluted to 1/100. After washes in cold PBS supplemented with 0.5% of bovine serum albumin (BSA), the cells were incubated for 30 minutes at 4°C with a goat anti-rabbit immunoglobulin-Alexa Fluor®488 diluted to 1/2000. After wash in cold PBS supplemented with 0.5% of BSA, the cells were analysed by C6 Flow Cytometer® (Accuri Cytometers, St. Ives, UK).

Hyaluronan receptor: CD44 receptor expression.

Flow cytometry were used to evaluate CD44 expression [45]. According to Pauloin’s method [23], cells were seeded into 6-well culture microplates, culture were kept at 37°C for 24 hours. The cells were incubated with different HA at 0.2% in culture medium with 2.5% of FCS. After 24 hours, culture medium was removed and the cells were washed and removed by trypsin. Culture cells were incubated for 45 minutes at 4°C with CD44 monoclonal antibodies (Sigma-Aldrich) diluted to 1/500. After three washes in cold PBS supplemented with 0.5% of BSA, the cells were incubated 30 minutes at 4°C with a goat anti-mouse immunoglobulin-FITC (Dakocytomation, Glostrup, Denmark) diluted to 1/2000. After three washes in cold PBS supplemented with 0.5% of BSA, the cells were fixed with 1% paraformaldehyde in PBS. The cells were analysed by C6 Flow Cytometer® (Accuri Cytometers, St. Ives, UK).

P2X7 receptor activation: YO-PRO-1 test.

YO-PRO-1, a DNA probe [46], enters cells after P2X7 receptor activation [13]. For this test, cells were seeded into 96-well culture microplates and kept at 37°C for 24 hours. The cells were incubated with different HA at 0.2% in culture medium with 2.5% of FCS. After 24 hours, culture medium was removed and the cells were washed with PBS. According to our laboratory’s protocol [47]; [48], 2 µM YO-PRO-1 solution was distributed in the microplate. After 10 minutes at room temperature in the dark, pictures of cells were taken using a Nikon Coolpix camera connected to a fluorescence microscope Leïca DMIRB.

Cell viability.

Neutral red (Fluka, Buchs, Switzerland) uptake assay is a cell viability assay, based on the ability of viable cells to incorporate neutral red [49], which is a weak cationic dye that readily penetrates cell membranes by non-ionic diffusion, accumulating in lysosomes, where it binds with anionic sites in the lysosomal matrix. Lysosomal membrane integrity is closely correlated with cell viability and it is evaluated with neutral red fluorescence. Cells were seeded into 96-well culture microplates, culture were kept at 37°C for 24 hours. The cells were incubated with different HA at 0.2% in culture medium with 2.5% of FCS. After 24 hours, culture medium was removed and the cells were washed. Neutral red solution (50 µg/ml) was added to living cells. After a 3-hour incubation time at 37°C, the cells were washed with PBS to remove any remaining unincorporated dye. The dye was then released from the cells using lysis solution (1% acetic acid, 50% ethanol and 49% pure water). The plate was agitated on a microplate shaker for 20 minutes and the fluorescence signal was scanned (λexc = 540 nm; λem = 600 nm) [50], using a cytofluorometer (Safire®, Tecan™, France) which allows fluorometric detection from 280 to 870 nm with high sensitivity (pg-fg/ml) and specificity. This technique allows the use of fluorescent probes on living cells and detects the fluorescent signal directly in the microplate in less than 1 minute (for a 96-well plate).

Statistical analysis.

Each test was performed in triplicate. The fluorescent measurement was expressed as relative fluorescence units or as fluorescence percentage of the control, and statistical analysis were performed using one-way ANOVA followed by Dunnett’s test (α risk = 0.05). Error bars on graphs represent standard deviation. The significance is compared to control (culture medium). Statistical analysis was performed using Sigma Stat 2.0 software (Chicago, IL, USA).

Acknowledgments

Authors would like to thank ADCIS for image analysis.

Author Contributions

Conceived and designed the experiments: KG PR. Performed the experiments: KG UD-P. Analyzed the data: KG J-MW PR. Wrote the paper: KG.

References

  1. 1. Weissman B, Meyer K (1954) The structure of hyalobiuronic acid and of hyaluronic acid from ombilical cord. J AM Chem Soc 27: 1753–1757.
  2. 2. Gandhi NS, Mancera RL (2008) The structure of glycosaminoglycans and their interactions with proteins. Chem Biol Drug Des 72: 455–482.
  3. 3. Laurent TC, Fraser JR (1992) Hyaluronan. Faseb J 6: 2397–2404.
  4. 4. Meyer K, Palmer J (1934) The polysaccahride of the vitreous humor. J Biol Chem 107: 620–634.
  5. 5. Fraser JR, Laurent TC, Laurent UB (1997) Hyaluronan: its nature, distribution, functions and turnover. J Intern Med 242: 27–33.
  6. 6. Chen WY, Abatangelo G (1999) Functions of hyaluronan in wound repair. Wound Repair Regen 7: 79–89.
  7. 7. Campo GM, Avenoso A, Campo S, D’Ascola A, Nastasi G, et al. (2010) Molecular size hyaluronan differently modulates toll-like receptor-4 in LPS-induced inflammation in mouse chondrocytes. Biochimie 92: 204–215.
  8. 8. Aruffo A, Stamenkovic I, Melnick M, Underhill CB, Seed B (1990) CD44 is the principal cell surface receptor for hyaluronate. Cell 61: 1303–1313.
  9. 9. Tammi R, MacCallum D, Hascall VC, Pienimaki JP, Hyttinen M, et al. (1998) Hyaluronan bound to CD44 on keratinocytes is displaced by hyaluronan decasaccharides and not hexasaccharides. J Biol Chem 273: 28878–28888.
  10. 10. Evanko SP, Angello JC, Wight TN (1999) Formation of hyaluronan- and versican-rich pericellular matrix is required for proliferation and migration of vascular smooth muscle cells. Arterioscler Thromb Vasc Biol 19: 1004–1013.
  11. 11. Nandi A, Estess P, Siegelman M (2004) Bimolecular complex between rolling and firm adhesion receptors required for cell arrest; CD44 association with VLA-4 in T cell extravasation. Immunity 20: 455–465.
  12. 12. Pike LJ, Han X, Gross RW (2005) Epidermal growth factor receptors are localized to lipid rafts that contain a balance of inner and outer leaflet lipids: a shotgun lipidomics study. J Biol Chem 280: 26796–26804.
  13. 13. Surprenant A, Rassendren F, Kawashima E, North RA, Buell G (1996) The cytolytic P2Z receptor for extracellular ATP identified as a P2X receptor (P2X7). Science 272: 735–738.
  14. 14. Takenouchi T, Sekiyama K, Sekigawa A, Fujita M, Waragai M, et al. (2010) P2X7 receptor signaling pathway as a therapeutic target for neurodegenerative diseases. Arch Immunol Ther Exp (Warsz) 58: 91–96.
  15. 15. Ballerini P, Rathbone MP, Di Iorio P, Renzetti A, Giuliani P, et al. (1996) Rat astroglial P2Z (P2X7) receptors regulate intracellular calcium and purine release. Neuroreport 7: 2533–2537.
  16. 16. Brandle U, Guenther E, Irrle C, Wheeler-Schilling TH (1998) Gene expression of the P2X receptors in the rat retina. Brain Res Mol Brain Res 59: 269–272.
  17. 17. MacKenzie AB, Surprenant A, North RA (1999) Functional and molecular diversity of purinergic ion channel receptors. Ann N Y Acad Sci 868: 716–729.
  18. 18. North RA, Surprenant A (2000) Pharmacology of cloned P2X receptors. Annu Rev Pharmacol Toxicol 40: 563–580.
  19. 19. Chiozzi P, Sanz JM, Ferrari D, Falzoni S, Aleotti A, et al. (1997) Spontaneous cell fusion in macrophage cultures expressing high levels of the P2Z/P2X7 receptor. J Cell Biol 138: 697–706.
  20. 20. Jiang LH, Mackenzie AB, North RA, Surprenant A (2000) Brilliant blue G selectively blocks ATP-gated rat P2X(7) receptors. Mol Pharmacol 58: 82–88.
  21. 21. Peng W, Cotrina ML, Han X, Yu H, Bekar L, et al. (2009) Systemic administration of an antagonist of the ATP-sensitive receptor P2X7 improves recovery after spinal cord injury. Proc Natl Acad Sci U S A 106: 12489–12493.
  22. 22. Pauloin T, Dutot M, Liang H, Chavinier E, Warnet JM, et al. (2009) Corneal protection with high-molecular-weight hyaluronan against in vitro and in vivo sodium lauryl sulfate-induced toxic effects. Cornea 28: 1032–1041.
  23. 23. Pauloin T, Dutot M, Warnet JM, Rat P (2008) In vitro modulation of preservative toxicity: high molecular weight hyaluronan decreases apoptosis and oxidative stress induced by benzalkonium chloride. Eur J Pharm Sci 34: 263–273.
  24. 24. Baricordi OR, Melchiorri L, Adinolfi E, Falzoni S, Chiozzi P, et al. (1999) Increased proliferation rate of lymphoid cells transfected with the P2X(7) ATP receptor. J Biol Chem 274: 33206–33208.
  25. 25. Mayo C, Steep MA, Trinkaus-Randal V (2007) The role of the P2X7 receptor in corneal wound healing. ARVO Annual Meeting 48: 3476.
  26. 26. Mankus C, Rich C, Minns M, Trinkaus-Randall V (2011) Corneal epithelium expresses a variant of P2X(7) receptor in health and disease. PLoS One 6: e28541.
  27. 27. Papakonstantinou E, Karakiulakis G, Roth M, Block LH (1995) Platelet-derived growth factor stimulates the secretion of hyaluronic acid by proliferating human vascular smooth muscle cells. Proc Natl Acad Sci USA 92: 9881–9885.
  28. 28. Moczar M, Robert L (1993) Stimulation of cell proliferation by hyaluronidase during in vitro aging of human skin fibroblasts. Exp Gerontol 28: 59–68.
  29. 29. Khaing ZZ, Milman BD, Vanscoy JE, Seidlits SK, Grill RJ, et al. (2011) High molecular weight hyaluronic acid limits astrocyte activation and scar formation after spinal cord injury. J Neural Eng 8: 046033.
  30. 30. Campo GM, Avenoso A, Campo S, D’Ascola A, Nastasi G, et al. (2010) Small hyaluronan oligosaccharides induce inflammation by engaging both toll-like-4 and CD44 receptors in human chondrocytes. Biochem Pharmacol 80: 480–490.
  31. 31. Steed E, Balda MS, Matter K (2010) Dynamics and functions of tight junctions. Trends Cell Biol 20: 142–149.
  32. 32. Brandner JM, Kief S, Grund C, Rendl M, Houdek P, et al. (2002) Organization and formation of the tight junction system in human epidermis and cultured keratinocytes. Eur J Cell Biol 81: 253–263.
  33. 33. Foger N, Marhaba R, Zoller M (2001) Involvement of CD44 in cytoskeleton rearrangement and raft reorganization in T cells. J Cell Sci 114: 1169–1178.
  34. 34. Ghatak S, Misra S, Toole BP (2005) Hyaluronan constitutively regulates ErbB2 phosphorylation and signaling complex formation in carcinoma cells. J Biol Chem 280: 8875–8883.
  35. 35. Adinolfi E, Callegari MG, Ferrari D, Bolognesi C, Minelli M, et al. (2005) Basal activation of the P2X7 ATP receptor elevates mitochondrial calcium and potential, increases cellular ATP levels, and promotes serum-independent growth. Mol Biol Cell 16: 3260–3272.
  36. 36. Bianco F, Ceruti S, Colombo A, Fumagalli M, Ferrari D, et al. (2006) A role for P2X7 in microglial proliferation. J Neurochem 99: 745–758.
  37. 37. Mayo C, Ren R, Rich C, Stepp MA, Trinkaus-Randall V (2008) Regulation by P2X7: epithelial migration and stromal organization in the cornea. Invest Ophthalmol Vis Sci 49: 4384–4391.
  38. 38. Gariboldi S, Palazzo M, Zanobbio L, Selleri S, Sommariva M, et al. (2008) Low molecular weight hyaluronic acid increases the self-defense of skin epithelium by induction of beta-defensin 2 via TLR2 and TLR4. J Immunol 181: 2103–2110.
  39. 39. Schroder JM, Harder J (1999) Human beta-defensin-2. Int J Biochem Cell Biol 31: 645–651.
  40. 40. Boukamp P, Petrussevska RT, Breitkreutz D, Hornung J, Markham A, et al. (1988) Normal keratinization in a spontaneously immortalized aneuploid human keratinocyte cell line. J Cell Biol 106: 761–771.
  41. 41. Todaro GJ, Lazar GK, Green H (1965) The initiation of cell division in a contact-inhibited mammalian cell line. J Cell Physiol 66: 325–333.
  42. 42. Liang CC, Park AY, Guan JL (2007) In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro. Nat Protoc 2: 329–333.
  43. 43. Fronza M, Heinzmann B, Hamburger M, Laufer S, Merfort I (2009) Determination of the wound healing effect of Calendula extracts using the scratch assay with 3T3 fibroblasts. J Ethnopharmacol 126: 463–467.
  44. 44. Russ PK, Davidson MK, Hoffman LH, Haselton FR (1998) Partial characterization of the human retinal endothelial cell tight and adherens junction complexes. Invest Ophthalmol Vis Sci 39: 2479–2485.
  45. 45. Sherman L, Skroch-Angel P, Moll J, Schwechheimer K, Ponta H, et al. (1995) Schwann cell tumors express characteristic patterns of CD44 splice variants. J Neurooncol 26: 171–184.
  46. 46. Idziorek T, Estaquier J, De Bels F, Ameisen JC (1995) YOPRO-1 permits cytofluorometric analysis of programmed cell death (apoptosis) without interfering with cell viability. J Immunol Methods 185: 249–258.
  47. 47. Dutot M, Pouzaud F, Larosche I, Brignole-Baudouin F, Warnet JM, et al. (2006) Fluoroquinolone eye drop-induced cytotoxicity: role of preservative in P2X7 cell death receptor activation and apoptosis. Invest Ophthalmol Vis Sci 47: 2812–2819.
  48. 48. Dutot M, Liang H, Pauloin T, Brignole-Baudouin F, Baudouin C, et al. (2008) Effects of toxic cellular stresses and divalent cations on the human P2X7 cell death receptor. Mol Vis 14: 889–897.
  49. 49. Borenfreund E, Puerner JA (1985) Toxicity determined in vitro by morphological alterations and neutral red absorption. Toxicol Lett 24: 119–124.
  50. 50. Rat P, Korwin-Zmijowska C, Warnet JM, Adolphe M (1994) New in vitro fluorimetric microtitration assays for toxicological screening of drugs. Cell Biol Toxicol 10: 329–337.