ORIGINAL ARTICLE

The impact of polyphenols on chondrocyte growth and survival: a preliminary report

Salvador Fernández-Arroyo1,2*, Fernando Huete-Toral3, María Jesús Pérez de Lara3, María de la Luz Cádiz-Gurrea4,5, Laurence Legeai-Mallet6, Vicente Micol7, Antonio Segura-Carretero4,5, Jorge Joven1,2 and Jesús Pintor3*

1Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, IISPV, Universitat Rovira i Virgili, Reus, Spain; 2Campus of International Excellence Southern Catalonia, Tarragona, Spain; 3Department of Biochemistry, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain; 4Functional Food Research and Development Center, Health Science Technological Park, Granada, Spain; 5Department of Analytical Chemistry, University of Granada, Granada, Spain; 6Imagine Institute – INSERM U1163, Necker Hospital for Sick Children, Paris, France; 7Institute of Molecular and Cell Biology, Miguel Hernández University, Elche, Spain

Abstract

Background: Imbalances in the functional binding of fibroblast growth factors (FGFs) to their receptors (FGFRs) have consequences for cell proliferation and differentiation that in chondrocytes may lead to degraded cartilage. The toxic, proinflammatory, and oxidative response of cytokines and FGFs can be mitigated by dietary polyphenols.

Objective: We explored the possible effects of polyphenols in the management of osteoarticular diseases using a model based on the transduction of a mutated human FGFR3 (G380R) in murine chondrocytes. This mutation is present in most cases of skeletal dysplasia and is responsible for the overexpression of FGFR3 that, in the presence of its ligand, FGF9, results in toxic effects leading to altered cellular growth.

Design: Different combinations of dietary polyphenols derived from plant extracts were assayed in FGFR3 (G380R) mutated murine chondrocytes, exploring cell survival, chloride efflux, extracellular matrix (ECM) generation, and grade of activation of mitogen-activated protein kinases.

Results: Bioactive compounds from Hibiscus sabdariffa reversed the toxic effects of FGF9 and restored normal growth, suggesting a probable translation to clinical requests in humans. Indeed, these compounds activated the intracellular chloride efflux, increased ECM generation, and stimulated cell proliferation. The inhibition of mitogen-activated protein kinase phosphorylation was interpreted as the main mechanism governing these beneficial effects.

Conclusions: These findings support the rationale behind the encouragement of the development of drugs that repress the overexpression of FGFRs and suggest the dietary incorporation of supplementary nutrients in the management of degraded cartilage.

Keywords: chondrodysplasia; fibroblast growth factor; Hibiscus sabdariffa; nutrition; osteoarthritis; polyphenols; signaling pathways

Citation: Food & Nutrition Research 2015, 59: 29311 - http://dx.doi.org/10.3402/fnr.v59.29311

Copyright: © 2015 Salvador Fernández-Arroyo et al. This is an Open Access article distributed under the terms of the Creative Commons Attribution 4.0 International License, allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material for any purpose, even commercially, provided the original work is properly cited and states its license.

Received: 30 July 2015; Revised: 8 September 2015; Accepted: 8 September 2015; Published: 5 October 2015

Conflict of interest and funding: The authors have no conflicts of interest.

*Correspondence to: Salvador Fernández-Arroyo, Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joanm, IISPV, Universitat Rovira i Virgili, C/ Sant Joan s/n, ES-43201 Reus, Spain, Email: sfernandez@fiispv.cat
*Jesús Pintor, Department of Biochemistry, Faculty of Optics and Optometry, Universidad Complutense de Madrid, C/Arcos de Jalón 118, ES-28037 Madrid, Spain, Email: jpintor@vet.ucm.es

To access the supplementary material to this article, please see Supplementary files under ‘Article Tools’.

 

The available evidence no longer sustains the concept that the biological activity of therapeutic compounds is limited to unique and well-defined mechanisms. Contrarily, the simultaneous and combined use of active constituents, which is in line with the notion that nutrients are associated with the preservation of health status, supports the challenging search for novel therapeutic targets among bioactive food components (1). In this setting, polyphenols associate their intrinsic antioxidant and anti-inflammatory properties with a synergistic impact in metabolic functions and maintenance of cellular homeostasis (2). The intimate mechanism(s) is not completely understood but is probably associated with gene expression pathways that coordinate important intracellular signaling pathways (3, 4). One of the most important ways to maintain correct cell growth and differentiation is via the signal transduction cascade, activated by the binding of growth factors to their receptors. In particular, fibroblast growth factors (FGFs) are crucial in the regulation of tissue formation (5), and the potential effects of altered interactions between FGFs and their receptors (FGFRs) are attractive targets to be explored in the relationship between food intake and disease (6).

In humans, the binding of growth factors to specific receptors results in a pleiotropic response derived from the distinct tissue distribution and the complexity of these families of proteins. However, their immediate action comes from the activation and/or dysfunction of phosphorylation events in important signaling pathways. Dietary polyphenols are probable candidates to modulate and regulate disturbances in these pathways through relevant key molecules, including phospholipase C gamma (PLCγ) (7), the signal transducer and activator of transcription (STAT) protein (8), phosphoinositol-3 kinase/protein kinase B (PI-3K/AKT) (9), and rat sarcoma protein/mitogen activated protein kinase (RAS/MAPK [ERK-1/-2]) (10). It is therefore expected that human nutrition and growth factors are associated with inflammatory and metabolic disturbances (1114). Moreover, point mutations in FGFRs affecting the function of FGFs-activated receptors have been shown to be consistently involved in cancer (1517), atherosclerosis (18), and dysfunctions in growth and development (19, 20).

For the purpose of this study, we highlight the fact that unambiguous functional mutations in FGFR3 are involved in several diseases (21, 22), including skeletal dysplasia and osteoarthritis (23, 24). Because dietary polyphenols block the actions of some cytokines and FGFs that lead to cartilage degradation (25), we have tested the hypothesis that dietary polyphenols might have an important regulatory effect on the functional binding of FGFs to their receptors. We used murine chondrocytes to transduce a human mutated FGFR3 by substituting glycine with arginine at position 380 (G380R), which causes achondroplasia. When the receptor is activated by its specific ligand (FGF9), this mutation produces severe changes in cellular growth. We have previously shown (2628) that this model is sensitive for gaining insight into the management of diseases associated with dysfunctions in cartilage. To reduce the complexities found in the composition of plant foods, but preserving the multiple and synergistic effects provided by the combination of multiple compounds, we have employed several fully characterized, polyphenol-rich plant extracts. The hypothesis is relevant because repression of mutated FGFR3 might illustrate the potential reversibility of a genetic condition and the feasibility of alternative therapeutic strategies (29, 30). Our findings support the view that the dietary incorporation of supplementary nutrients may be safely utilized to regulate intracellular signaling networks in the management of degraded cartilage phenotypes.

Material and methods

Chemical and reagents

Tetracycline, minimum essential medium alpha (α-MEM), heat-inactivated fetal bovine serum, and antibiotics (penicillin, Geneticin, streptomycin, and hygromycin) were obtained from Invitrogen (Carlsbad, CA, USA). Tris base, NaCl, NP-40, sodium deoxycholate, SDS, TWEEN-20, BSA, phenylmethylsulfonyl fluoride (PMSF), sodium fluoride (NaF), sodium orthovanadate (Na3VO4), aprotinin, pepstatin, leupeptin, and human recombinant FGF9 were obtained from Sigma (St. Louis, MO, USA). Antibodies against phospho-ERK-1/-2, ERK-1/-2, and GAPDH as well as horseradish peroxidase-conjugated goat anti-mouse IgG were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA).

Plant extracts and phenolic composition

We assayed different combinations of polyphenols in extracts prepared at a final concentration of 100 µg/mL in water or dimethyl sulfoxide from Aspalathus linearis (family Fabaceae; rooibos leaves), Vitis vinifera (family Vitaceae; grape seeds), Citrus aurantium (family Rutaceae; bitter orange), Lippia citriodora (family Verbenaceae; lemon verbena leaves), Olea europaea (family Oleaceae; olive leaves), and Hibiscus sabdariffa (family Malvaceae; karkadé). The criteria for inclusion were as follows: previous use in our laboratory in cell or animal models, commercialization (usually to prepare beverages), chemical characterization, and safety for human consumption. All extracts are readily available (Monteloeder, Elche, Spain) and information on the composition is provided in Supplementary Tables 1 and 2. Chromatographic methods, along with any additional extended data and references unique to these sections, are available as supplementary information in the online version. Of note, we also tested extracts from Hypoxis rooperi (family Hypoxidaceae; African potato) to ensure the lack of effects from lignans (data not shown). Because of distinct and significant effects obtained from H. sabdariffa, we only present a full report of data obtained with this mixture and a new preparation to concentrate its phenolic compounds. This concentration was performed via a described procedure that eliminates the residual presence of sugar, fiber, and other material, significantly reducing the concentration of organic acids and decreasing the concentration of prodelphinidin B3 (31) (Supplementary Table 2). This concentrated extract was employed in subsequent experiments to provide a concentration of compounds similar to that after the addition of normal extract (10 µg/mL in the culture medium).

Cell culture and assessment of cell size and morphology

RCJ3.1C5.18 cells obtained from a mesenchymal cell line that differentiates into chondrocytes were used to test whether polyphenols modulate the FGF system. Cells were stably transfected with full-length human wild type (RCJ-FGFR3-WT) cells or with the G380R mutant FGFR3 (RCJ-FGFR3-G380R) cells included in a vector that overexpresses the receptor in absence of tetracycline, following procedures previously described (32, 33). Mutated FGFR3 (G380R) results in the overexpression of FGFR3, and in this model FGF9 provokes a reduction in cell growth and aberrant morphological changes in chondrocytes. The required materials were obtained from ProChon Biotech (Woburn, MA, USA). Cells were incubated at 37°C with 5% CO2 in the standard α-MEM culture medium supplemented with 15% heat-inactivated fetal bovine serum and antibiotics (600 µg/mL Geneticin, 100 U/mL penicillin/streptomycin, and 50 µg/mL hygromycin) with or without 2 µg/mL tetracycline. Results were provided by six independent experiments, in triplicate, and trypan blue was employed to detect viable, unstained cells. Cell size and morphology were monitored microscopically, as previously described (28).

Chloride efflux measurements

To explore the effects of FGF9 in morphologic changes, we first measured the FGFR3-regulated chloride flux to assess the role of voltage gated chloride channels in chondrogenesis (34). This method was employed as a screening procedure to assess the potential benefits provided by polyphenols. In brief, measurements were made using N-[ethoxycarbonylmethyl]-6-methoxy-quinolinium bromide (MQAE) as a fluorescent indicator (35). MQAE has a high sensitivity to chloride, and changes in fluorescence inversely reflect changes in intracellular chloride concentration. Thus, a decrease in intracellular fluorescence indicates the conservation of or an increase in the intracellular chloride concentration. In the same way, an increase in fluorescence indicates a decrease in the intracellular chloride concentration. In both cases, a significant movement of water is produced with a consequent change in the voltage of the membrane (36). As previously published (28), measurements were made employing the appropriate controls, and the final concentration of FGF9 was set at 25 ng/mL according to dose–response experiments. Cells were grown to confluence in 96-well plates and loaded overnight with 0.8 mM MQAE at 37°C. Then, the medium was removed, washed, and incubated with chloride-containing buffer (to induce chloride channel activation) for 10 min at 37°C. The buffer was then removed and replaced by 100 µL of chloride-free buffer containing the desired concentration of FGF9.

Assessment of ERK-1/-2 phosphorylation

Signals evoked by ligand-receptor interactions control fundamental cellular processes in chondrocytes. The experiments were focused to assess the previously unexplored effects of H. sabdariffa on the ERK-1 and -2 components of the MAPK cascade. ERK-1/-2 activation would represent an induction of chondrocyte maturation and, conversely, inhibition of ERK-1/-2 would suggest a blockade of maturation and cell death (37). Cells were lysed in a buffer with the following composition: 50 mM Tris–HCl pH 8.0, 150 mM NaCl, 1% NP-40, 0.5% sodium deoxycholate, 0.1% SDS, 1 mM PMSF, 1 mM NaF, 2 mM Na3VO4, 10 µg/mL aprotinin, 5 µg/mL pepstatin, and 10 µg/mL leupeptin. After centrifugation at 13,000 g for 20 min at 4°C, protein concentration was determined by the Bio-Rad protein assay (Bio-Rad laboratories, Hercules, CA, USA). Proteins from each sample (45 µg) were subjected to 10% SDS-polyacrylamide gels and were transferred to nitrocellulose membranes (Amersham Pharmacia Biotech, Buckinghamshire, UK). Membranes were then blocked and incubated overnight with the primary antibody in TBS containing 5% skim milk and 0.1% TWEEN-20. After washing, blots were incubated with horseradish peroxidase-conjugated goat anti-mouse IgG secondary antibody (1:2,000). Development was performed utilizing the ECL system (Amersham Pharmacia Biotech). Films were scanned and analyzed using a Kodak GL 200 Imaging System with Kodak Molecular Imaging Software (Rochester, NY, USA). Data are the result of six independent experiments, in triplicate, and the overall procedure was assessed using GAPDH as control.

Measurement of extracellular matrix secretion

The extracellular matrix (ECM) provides mechanical support and a spatial context for signaling events by various cell surface growth factor receptors (38). We studied the effects of H. sabdariffa extract and concentrated polyphenols in cartilage matrix deposition in chondrocytes with a method based on Alcian blue staining (39). In brief, reagents and protocols were obtained from Lifeline Cell Technology (Frederick, MD, USA) and PromoCell (Heidelberg, Germany). Cells were seeded at a density of 2×105 cells/well in six-well dishes, and differentiation was induced by adding 10 mM β-glycerophosphate and 50 µg/mL ascorbic acid to the medium. Cultures were fed with supplemented media every 2 days; FGF9 (25 ng/mL) was added to fresh growth medium, alone or in combination with bioactive compounds. Under these conditions, the amount of cartilage matrix (proteoglycan synthesis) was measured at 3, 7, and 10 days of culture (39). At these time points, cells were washed with PBS and stained with Alcian blue stain (1% in 3% acetic acid) for 30 min, washed again three times for 2 min in 3% acetic acid, and rinsed with distilled water. Solubility was achieved with 1% SDS, 1 h at 90°C, to measure the absorbance at 605 nm.

Statistical analysis

All data are presented as the mean±SD. To test general differences among means, we employed ANOVA, followed by the Tukey-Kramer post-hoc test. To test specific differences between means, homogenous variances were assumed by the F-test, and Student’s t-test (two-sided) was employed. P<0.05 was considered to be statistically significant. Graphics were prepared with GraphPad Prism 6 (La Jolla, CA, USA) in combination with the statistical software SPSS version 19 (Chicago, IL, USA).

Results and discussion

The expression in chondrocytes of the FGFR3 (G380R) mutation is currently considered to be the likely cause of the human chondrodysplasia, termed achondroplasia(40, 41). In these patients, there is an over-activation of FGFR3 in the presence of FGF9, which results in toxic effects in chondrocytes, preventing endochondral bone formation. Because this represents a gain-of-function mutation and FGFR3 is a transmembrane receptor tyrosine kinase that initiates signal transduction to the nucleus, it is reasonable to predict that successful therapeutic strategies would decrease or eliminate FGFR3 signals. Using mouse chondrocytes containing the human mutant FGFR3, it has been possible to test the effects of polyphenols and how these and other plant dietary compounds affect intracellular signaling networks in an achondroplastic model (2). To follow this approach, we measured the effect of commercially available extracts commonly used in edible preparations on the modulation of chloride efflux by FGFR3 (G380R) and FGF9. The arrangement of FGF9 and FGFR3 in this model induced an enlarged achondroplastic chondrocyte size and an increase in the intracellular chloride, which is likely the result of an inhibition of the chloride efflux (42). Intriguingly, most polyphenol combinations further increased the intracellular chloride concentration and potentiated the deleterious effects in chondrocytes (Fig. 1a). Quantitative effects, however, differed considerably, and we noted significant changes in cell size and a disproportionate 15-fold increase in intracellular chloride concentration caused by C. aurantium. In response to the reproducibility of these data, we excluded these dietary extracts in this study, but this information will drive studies to further investigate the role of resting membrane potential in the normal development of chondrocytes. Conversely, compounds from H. sabdariffa restrained the increase in the intracellular chloride concentration (Fig. 1b). Interpretation is difficult because data could indicate combined actions on FGFR3 signaling, binding of FGF9, or direct preservation of the membrane potential. Furthermore, analysis of the composition of the complex mixture of compounds in the initial extract revealed that a significant amount of organic acids and other materials were present together with polyphenols. We then eliminated these residual components to obtain an extract in which polyphenols were concentrated. Subsequent experiments, once corrected for the total amount of compounds, indicated that the effect of polyphenols in the regulation of chloride efflux, although significant, was less intense than that observed in the initial extract (Fig. 1b and Supplementary Tables 1 and 2). Contrary to what we expected, the results suggest a major role for organic acids, particularly hydroxycitric acid, but attribution of the effect to a single component is highly unlikely. Moreover, when we utilized both mixtures derived from H. sabdariffa, polyphenols appeared to be the compounds causing the inhibition of ERK-1/-2 (p42 and p44) phosphorylation. ERKs are important in cellular growth and development (17, 4348), and the effects were detectable in cultures without FGF9, but statistical significance was only achieved in the presence of the ligand (Fig. 2). This effect is relevant because ERK-1/-2 phosphorylation was activated in FGFR3 (G380R) chondrocytes challenged with FGF9, confirming our previous findings (28). The role of the original H. sabdariffa extract may not be inferred from our data because it had no significant effect on the level of phosphorylation with or without FGF9, but it has been shown to target other multiple components (4954). However, we limited the exploration to the MAPK pathway because ERK-1/-2 inhibition under these conditions is probably restricted to chondrocytes, in which this signaling system regulates cell growth, proliferation, and ECM accumulation (5562).

Fig 1

Fig. 1.   Intracellular chloride accumulation is modulated by polyphenols. Measurements are indicated as percentages with respect to values obtained in wild type cells and the effect of the addition of FGF9 (25 ng/mL; 100%) or the respective controls without FGF9 (0%). Data are expressed as the mean±SD (n=6–9 independent experiments in triplicate), indicating the relative increase or decrease in chloride concentration. (a) Among the different combinations of polyphenols, only those provided by Hibiscus sabdariffa decreased intracellular chloride accumulation. In contrast, all other combinations favored the accumulation of chloride. We decided to exclude these extracts in further analyses, and p values were provided only for H. Sabdariffa. (b) We concentrated this extract to individualize the effect of polyphenols, assessing any potential contribution of residual compounds and organic acids, and the values were compared with those obtained from the initial H. sabdariffa extract. Asterisks denote significant effects with respect to controls.

Fig 2

Fig. 2.   Relative ERK-1/-2 phosphorylation. Experiments were performed in FGFR3 (G380R) mutant RCJ3.1C5.18 chondrocytes with or without FGF9 (25 ng/mL) and further incubated with Hibiscus sabdariffa or its polyphenolic extract. Data were combined and expressed as the mean±SD (n=6, in triplicate). Representative results were included for illustrative purposes.

The stimulation by FGF9 of mutated FGFR3 also results in a dramatic decrease in ECM production in chondrocytes, an action that was reversed by H. sabdariffa. The effect was ostensible even in the absence of FGF9 and reached statistical significance after 10 days of treatment (Fig. 3). Total values were lower than those observed in wild type cells, indicating the probable contribution of other mechanisms, but active studies suggest that the differences are reduced with longer exposure time and higher concentrations (data not shown). ECM enrichment improves chondrocyte physiology due to the optimization of the signaling microenvironment of FGFs (63, 64). This effect is relevant in this environment because the formation of molecular complexes between growth factors and ECM proteins regulates dynamic interactions between proteins, bioavailability and function of many growth factors, and the modulation of FGFR signaling (6567). It is therefore plausible that, in skeletal dysplasia and in diseases with prominently degraded cartilage, FGF9/FGFR3 modulators could prevent cartilage degeneration and/or promote cartilage regeneration (68, 69).

Fig 3

Fig. 3.   Extracellular matrix (ECM) deposition. Data were combined and expressed as the mean±SD (n=6, in triplicate), and the values indicate the amount of enrichment in the ECM utilizing Alcian blue staining of cells. Measurements were sequentially performed at days 3, 7, and 10 and included untreated cells (control) and activated cells in the presence of Hibiscus sabdariffa extract or its derived polyphenolic extract with or without FGF9 (25 ng/mL).

Accordingly, chondrocyte viability is low when cells bear the (G380R) mutation of FGFR3, the likely consequence of increased maturation and early cell death (70, 71). The effect is highly dependent on the presence of the ligand (FGF9), indicating the importance of active growth factors in the vicinity. In this model, FGF9 decreased cell survival in a few hours. H. sabdariffa treatment completely reversed the action of FGF9, and cell proliferation was similar to that observed in controls (Fig. 4), indicating the rescue of the FGFR3 (G380R) phenotype. Experiments should follow to ascertain whether individual compounds may elicit similar beneficial effects. However, safe concentrations are more difficult to achieve, and observations in animal models suggest that the synergistic action of natural compounds is a more likely possibility (3, 31, 55). Stronger evidence could be provided by the development of better in vivo and in vitro models employing human cells. It should be mentioned that recent cell reprogramming technologies might be utilized to obtain induced pluripotent stem cells differentiated into mutant chondrocytes that may confirm the anabolic effects we have found in chondrocytes treated with bioactive compounds from H. sabdariffa. As preliminary results, immunoprecipitation experiments of FGFR3 suggest that these bioactive compounds are not affecting the receptor directly. Thus, the exploration of related signaling pathways other than ERK-1/-2 in chondrocytes and the effect of some available drugs might also contribute to fully uncovering the actual molecular mechanisms (7274). Perhaps it is time to contemplate significant dietary modifications in humans by food design and supplementation to obtain health benefits (75).

Fig 4

Fig. 4.   Chondrocyte proliferation comparing untreated cells (control) and treated cells with Hibiscus sabdariffa and its polyphenolic extract with or without FGF9 (25 ng/mL). Cell viability was determined by the trypan blue method. Data are expressed as mean±SD (n=6, in triplicate).

Conclusion

With the rationale that some nutrients with intrinsic anti-inflammatory and antioxidative effects may mitigate a deleterious response leading to degraded cartilage, we assayed the activity of dietary polyphenols in murine cells with the transduced human (G380R) mutation in FGFR3. Most polyphenols had no effect, but those provided by H. sabdariffa rescued chondrocytes from FGF9-induced toxic effects, supporting the view that plant-derived dietary products affect intracellular signaling networks and may be safely utilized in the management of degraded cartilage phenotypes. H. sabdariffa rescued mutated FGFR3 chondrocytes, restoring normal growth, decreasing the intracellular chloride concentration, inhibiting ERK-1/-2 phosphorylation, and increasing the generation of ECM (Fig. 5). Our data indicate the crucial role of growth factors in the survival of chondrocytes and suggest effective and applicable therapeutic strategies for patients with degraded cartilage. H. sabdariffa would have the advantage that it has been safely administered to large numbers of humans for millennia, including pregnant women and infants.

Fig 5

Fig. 5.   Illustrative summary of observed and proposed effects in the mutant chondrocyte model. Polyphenols target multiple components that promote and maintain tissue homeostasis beyond their anti-inflammatory and antioxidant effects, and their actions might be extended to disease prevention and intervention. Binding of fibroblast growth factor (FGF9) to its receptor (FGFR3) activates several important signaling pathways that are associated with the overall response to inflammation and are crucial in chondrocytes to regulate cell proliferation and extracellular matrix (ECM) generation. From the data, we consider the possibility that the action of complete Hibiscus sabdariffa extract might be different from that observed with concentrated polyphenols. In particular, polyphenols appear more active in the inhibition of ERK-1/-2 phosphorylation and in the increase of ECM generation, whereas the combination of organic acids and polyphenols could be more active in reversing the intracellular chloride accumulation and increasing cellular proliferation. H. sabdariffa reverses the toxic actions of the FGF9/FGFR3 interaction in mutated chondrocytes. The results indicate the need for a search for human models to fully establish actual mechanisms as well as the potential modification of diets to achieve human health benefits. AKT (PKB): protein kinase B; ERK 1/2 (MAPK): extracellular signal-regulated kinases 1 and 2; FGF: fibroblast growth factor; FGFR: fibroblast growth factor receptor; MEK1/2: MAPK kinases 1 and 2; PI-3K: phosphatidylinositide 3-kinase; PLCγ: phospholipase C gamma; RAF: rapidly accelerated fibrosarcoma (serine/threonine-protein kinase); RAS: rat sarcoma protein (GTPase); STAT1: signal transducer and activator of transcription 1.

Acknowledgements

The authors are grateful for the constant support provided by the Hospital Universitari de Sant Joan and the Universitat Rovira i Virgili. Salvador Fernández-Arroyo is the recipient of a Sara Borrell grant (CD12/00672) from the Instituto de Salud Carlos III, Madrid, Spain. The authors also thank the Andalusian Regional Government Council of Innovation and Science for the Excellence Project P11-CTS-7625 and Generalitat Valenciana for the project PROMETEO/2012/007. This work was also supported by projects of the Fundación Areces and the Fundación MAGAR. These institutions had no involvement in the study design, analysis, or interpretation of data, writing of the report, or in the decision to submit the article for publication.

References

  1. Menendez JA, Joven J, Aragones G, Barrajon-Catalan E, Beltran-Debon R, Borras-Linares I, et al. Xenohormetic and anti-aging activity of secoiridoid polyphenols present in extra virgin olive oil: a new family of gerosuppressant agents. Cell Cycle 2013; 12: 555–78. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  2. Joven J, Rull A, Rodriguez-Gallego E, Camps J, Riera-Borrull M, Hernandez-Aguilera A, et al. Multifunctional targets of dietary polyphenols in disease: a case for the chemokine network and energy metabolism. Food Chem Toxicol 2013; 51: 267–79. PubMed Abstract | Publisher Full Text
  3. Joven J, Espinel E, Rull A, Aragones G, Rodriguez-Gallego E, Camps J, et al. Plant-derived polyphenols regulate expression of miRNA paralogs miR-103/107 and miR-122 and prevent diet-induced fatty liver disease in hyperlipidemic mice. Biochim Biophys Acta 2012; 1820: 894–9. PubMed Abstract | Publisher Full Text
  4. Joven J, Micol V, Segura-Carretero A, Alonso-Villaverde C, Menendez JA, Platform BFC. Polyphenols and the modulation of gene expression pathways: can we eat our way out of the danger of chronic disease? Crit Rev Food Sci Nutr 2014; 54: 985–1001. PubMed Abstract | Publisher Full Text
  5. Takei Y, Minamizaki T, Yoshiko Y. Functional diversity of fibroblast growth factors in bone formation. Int J Endocrinol 2015; 2015: 729352. PubMed Abstract | PubMed Central Full Text
  6. Lelliott CJ, Ahnmark A, Admyre T, Ahlstedt I, Irving L, Keyes F, et al. Monoclonal antibody targeting of fibroblast growth factor receptor 1c ameliorates obesity and glucose intolerance via central mechanisms. PLoS One 2014; 9: e112109. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  7. Abe M, Naski MC. Regulation of sprouty expression by PLC-gamma and calcium-dependent signals. Biochem Biophys Res Comm 2004; 323: 1040–7. PubMed Abstract | Publisher Full Text
  8. Hart KC, Robertson SC, Donoghue DJ. Identification of tyrosine residues in constitutively activated fibroblast growth factor receptor 3 involved in mitogenesis, stat activation, and phosphatidylinositol 3-kinase activation. Mol Biol Cell 2001; 12: 931–42. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  9. d’Anglemont de Tassigny A, Berdeaux A, Souktani R, Henry P, Ghaleh B. The volume-sensitive chloride channel inhibitors prevent both contractile dysfunction and apoptosis induced by doxorubicin through PI3kinase, Akt and Erk 1/2. Eur J Heart Fail 2008; 10: 39–46. PubMed Abstract | Publisher Full Text
  10. Cross MJ, Lu L, Magnusson P, Nyqvist D, Holmqvist K, Welsh M, et al. The Shb adaptor protein binds to tyrosine 766 in the FGFR-1 and regulates the Ras/MEK/MAPK pathway via FRS2 phosphorylation in endothelial cells. Mol Biol Cell 2002; 13: 2881–93. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  11. Daleprane JB, Freitas Vda S, Pacheco A, Rudnicki M, Faine LA, Dorr FA, et al. Anti-atherogenic and anti-angiogenic activities of polyphenols from propolis. J Nutr Biochem 2012; 23: 557–66. PubMed Abstract | Publisher Full Text
  12. Sarruf DA, Thaler JP, Morton GJ, German J, Fischer JD, Ogimoto K, et al. Fibroblast growth factor 21 action in the brain increases energy expenditure and insulin sensitivity in obese rats. Diabetes 2010; 59: 1817–24. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  13. Tanaka T, Ngwa JS, van Rooij FJ, Zillikens MC, Wojczynski MK, Frazier-Wood AC, et al. Genome-wide meta-analysis of observational studies shows common genetic variants associated with macronutrient intake. Am J Clin Nutr 2013; 97: 1395–402. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  14. Wu J, Wei T, Tang Q, Weng B, Li W, Jiang X, et al. Discovery and anti-cancer evaluation of two novel non-ATP-competitive FGFR1 inhibitors in non-small-cell lung cancer. BMC Cancer 2015; 15: 276. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  15. Kelleher FC, O’Sullivan H, Smyth E, McDermott R, Viterbo A. Fibroblast growth factor receptors, developmental corruption and malignant disease. Carcinogenesis 2013; 34: 2198–205. PubMed Abstract | Publisher Full Text
  16. Lemmon MA, Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell 2010; 141: 1117–34. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  17. Song JY, Kim CS, Lee JH, Jang SJ, Lee SW, Hwang JJ, et al. Dual inhibition of MEK1/2 and EGFR synergistically induces caspase-3-dependent apoptosis in EGFR inhibitor-resistant lung cancer cells via BIM upregulation. Invest New Drugs 2013; 31: 1458–65. PubMed Abstract | Publisher Full Text
  18. Raj T, Kanellakis P, Pomilio G, Jennings G, Bobik A, Agrotis A. Inhibition of fibroblast growth factor receptor signaling attenuates atherosclerosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 2006; 26: 1845–51. PubMed Abstract | Publisher Full Text
  19. Bohensky J, Terkhorn SP, Freeman TA, Adams CS, Garcia JA, Shapiro IM, et al. Regulation of autophagy in human and murine cartilage hypoxia-inducible factor 2 suppresses chondrocyte autophagy. Arthritis Rheum 2009; 60: 1406–15. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  20. Lazarus JE, Hegde A, Andrade AC, Nilsson O, Baron J. Fibroblast growth factor expression in the postnatal growth plate. Bone 2007; 40: 577–86. PubMed Abstract | Publisher Full Text
  21. Al Hussain TO, Akhtar M. Molecular basis of urinary bladder cancer. Adv Anat Pathol 2013; 20: 53–60. PubMed Abstract | Publisher Full Text
  22. Blomberg M, Jeppesen EM, Skovby F, Benfeldt E. FGFR3 mutations and the skin: report of a patient with a FGFR3 gene mutation, acanthosis nigricans, hypochondroplasia and hyperinsulinemia and review of the literature. Dermatology 2010; 220: 297–305. PubMed Abstract | Publisher Full Text
  23. Baujat G, Legeai-Mallet L, Finidori G, Cormier-Daire V, Le Merrer M. Achondroplasia. Best Pract Res Clin Rheumatol 2008; 22: 3–18. PubMed Abstract | Publisher Full Text
  24. Foldynova-Trantirkova S, Wilcox WR, Krejci P. Sixteen years and counting: the current understanding of fibroblast growth factor receptor 3 (FGFR3) signaling in skeletal dysplasias. Hum Mutat 2012; 33: 29–41. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  25. Leong DJ, Choudhury M, Hirsh DM, Hardin JA, Cobelli NJ, Sun HB. Nutraceuticals: potential for chondroprotection and molecular targeting of osteoarthritis. Int J Mol Sci 2013; 14: 23063–85. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  26. Guzman-Aranguez A, Irazu M, Yayon A, Pintor J. Effect of diadenosine polyphosphates in achondroplasic chondrocytes: inhibitory effect of Ap(4)A on FGF9 induced MAPK cascade. Biochem Pharmacol 2007; 74: 448–56. PubMed Abstract | Publisher Full Text
  27. Guzman-Aranguez A, Irazu M, Yayon A, Pintor J. P2Y receptors activated by diadenosine polyphosphates reestablish Ca2+ transients in achondroplasic chondrocytes. Bone 2008; 42: 516–23. PubMed Abstract | Publisher Full Text
  28. Huete F, Guzman-Aranguez A, Ortin J, Hoyle CHV, Pintor J. Effects of diadenosine tetraphosphate on FGF9-induced chloride flux changes in achondroplastic chondrocytes. Purinergic Signal 2011; 7: 243–9. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  29. Garcia S, Dirat B, Tognacci T, Rochet N, Mouska X, Bonnafous S, et al. Postnatal soluble FGFR3 therapy rescues achondroplasia symptoms and restores bone growth in mice. Sci Transl Med 2013; 5: 203ra124. PubMed Abstract
  30. Oliveras-Ferraros C, Cufi S, Queralt B, Vazquez-Martin A, Martin-Castillo B, de Llorens R, et al. Cross-suppression of EGFR ligands amphiregulin and epiregulin and de-repression of FGFR3 signalling contribute to cetuximab resistance in wild-type KRAS tumour cells. Br J Cancer 2012; 106: 1406–14. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  31. Herranz-Lopez M, Fernandez-Arroyo S, Perez-Sanchez A, Barrajon-Catalan E, Beltran-Debon R, Abel Menendez J, et al. Synergism of plant-derived polyphenols in adipogenesis: perspectives and implications. Phytomedicine 2012; 19: 253–61. PubMed Abstract | Publisher Full Text
  32. Monsonego-Ornan E, Adar R, Feferman T, Segev O, Yayon A. The transmembrane mutation G380R in fibroblast growth factor receptor 3 uncouples ligand-mediated receptor activation from down-regulation. Mol Cell Biol 2000; 20: 516–22. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  33. Monsonego-Ornan E, Adar R, Rom E, Yayon A. FGF receptors ubiquitylation: dependence on tyrosine kinase activity and role in downregulation. FEBS Lett 2002; 528: 83–9. PubMed Abstract | Publisher Full Text
  34. Tian M, Duan Y, Duan X. Chloride channels regulate chondrogenesis in chicken mandibular mesenchymal cells. Arch Oral Biol 2010; 55: 938–45. PubMed Abstract | Publisher Full Text
  35. Koncz C, Daugirdas JT. Use of MQAE for measurement of intracellular [Cl-] in cultured aortic smooth muscle cells. Am J Physiol 1994; 267: H2114–23. PubMed Abstract
  36. Marandi N, Konnerth A, Garaschuk O. Two-photon chloride imaging in neurons of brain slices. Pflugers Arch 2002; 445: 357–65. PubMed Abstract | Publisher Full Text
  37. Yoon YM, Kim SJ, Oh CD, Ju JW, Song WK, Yoo YJ, et al. Maintenance of differentiated phenotype of articular chondrocytes by protein kinase C and extracellular signal-regulated protein kinase. J Biol Chem 2002; 277: 8412–20. PubMed Abstract | Publisher Full Text
  38. Kim SH, Turnbull J, Guimond S. Extracellular matrix and cell signalling: the dynamic cooperation of integrin, proteoglycan and growth factor receptor. J Endocrinol 2011; 209: 139–51. PubMed Abstract | Publisher Full Text
  39. Lunstrum GP, Keene DR, Weksler NB, Cho YJ, Cornwall M, Horton WA. Chondrocyte differentiation in a rat mesenchymal cell line. J Histochem Cytochem 1999; 47: 1–6. PubMed Abstract | Publisher Full Text
  40. Rousseau F, Bonaventure J, Legeai-Mallet L, Pelet A, Rozet JM, Maroteaux P, et al. Mutations in the gene encoding fibroblast growth factor receptor-3 in achondroplasia. Nature 1994; 371: 252–4. PubMed Abstract | Publisher Full Text
  41. Warman ML, Cormier-Daire V, Hall C, Krakow D, Lachman R, LeMerrer M, et al. Nosology and classification of genetic skeletal disorders: 2010 revision. Am J Med Genet A 2011; 155a: 943–68. PubMed Abstract | Publisher Full Text
  42. Lewis R, Asplin KE, Bruce G, Dart C, Mobasheri A, Barrett-Jolley R. The role of the membrane potential in chondrocyte volume regulation. J Cell Physiol 2011; 226: 2979–86. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  43. Britten CD. PI3K and MEK inhibitor combinations: examining the evidence in selected tumor types. Cancer Chemother Pharmacol 2013; 71: 1395–409. PubMed Abstract | Publisher Full Text
  44. Dent P. ERK plays the baddie (again). Cancer Biol Ther 2013; 14: 997–8. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  45. Dhawan V, Mahajan N, Bhat OM, Bahl A, Sharma I. ERK signaling is involved in inflammation and vascular remodeling process in premature coronary artery disease. Atheroscler Suppl 2011; 12: 88. Publisher Full Text
  46. Johnson GL, Nakamura K. The c-jun kinase/stress-activated pathway: regulation, function and role in human disease. Biochim Biophys Acta 2007; 1773: 1341–8. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  47. Lesch ME, Huber C, Jordan HJ, Schrier D, Thiel M, Flory C. The effect of inhibiting the MEK-ERK MAPK pathway on inflammation and articular cartilage integrity in a rat model of collagen-induced arthritis. FASEB J 2001; 15: A1061.
  48. Saini KS, Loi S, de Azambuja E, Metzger-Filho O, Saini ML, Ignatiadis M, et al. Targeting the PI3K/AKT/mTOR and Raf/MEK/ERK pathways in the treatment of breast cancer. Cancer Treat Rev 2013; 39: 935–46. PubMed Abstract | Publisher Full Text
  49. Ajiboye TO, Salawu NA, Yakubu MT, Oladiji AT, Akanji MA, Okogun JI. Antioxidant and drug detoxification potentials of Hibiscus sabdariffa anthocyanin extract. Drug Chem Toxicol 2011; 34: 109–15. PubMed Abstract | Publisher Full Text
  50. Beltran-Debon R, Alonso-Villaverde C, Aragones G, Rodriguez-Medina I, Rull A, Micol V, et al. The aqueous extract of Hibiscus sabdariffa calices modulates the production of monocyte chemoattractant protein-1 in humans. Phytomedicine 2010; 17: 186–91. PubMed Abstract | Publisher Full Text
  51. Fernandez-Arroyo S, Camps J, Menendez JA, Joven J. Managing hypertension by polyphenols. Planta Med 2015; 81: 624–9. PubMed Abstract | Publisher Full Text
  52. Lin HH, Chen JH, Wang CJ. Chemopreventive properties and molecular mechanisms of the bioactive compounds in Hibiscus sabdariffa Linne. Curr Med Chem 2011; 18: 1245–54. PubMed Abstract | Publisher Full Text
  53. Ubani CS, Joshua PE, Anieke UC. Effects of aqueous extract of Hibiscus sabdariffa L. calyces on liver marker enzymes of phenobarbitone-induced adult wistar albino rats. J Chem Pharm Res 2011; 3: 528–37.
  54. Yang MY, Peng CH, Chan KC, Yang YS, Huang CN, Wang CJ. The hypolipidemic effect of Hibiscus sabdariffa polyphenols via inhibiting lipogenesis and promoting hepatic lipid clearance. J Agric Food Chem 2010; 58: 850–9. PubMed Abstract | Publisher Full Text
  55. Harada D, Yamanaka Y, Ueda K, Nishimura R, Morishima T, Seino Y, et al. Sustained phosphorylation of mutated FGFR3 is a crucial feature of genetic dwarfism and induces apoptosis in the ATDC5 chondrogenic cell line via PLC gamma-activated STATI. Bone 2007; 41: 273–81. PubMed Abstract | Publisher Full Text
  56. Im HJ, Li X, Chen D, Yan D, Kim J, Ellman MB, et al. Biological effects of the plant-derived polyphenol resveratrol in human articular cartilage and chondrosarcoma cells. J Cell Physiol 2012; 227: 3488–97. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  57. Kim SJ, Kim HG, Oh CD, Hwang SG, Song WK, Yoo YJ, et al. p38 kinase-dependent and -independent Inhibition of protein kinase C zeta and -alpha regulates nitric oxide-induced apoptosis and dedifferentiation of articular chondrocytes. J Biol Chem 2002; 277: 30375–81. PubMed Abstract | Publisher Full Text
  58. Lee WK, Chung KW, Kim GH, Kim SJ. Gallotannin causes differentiation and inflammation via ERK1/2 and p38 kinase pathways in rabbit articular chondrocytes. Mol Med Rep 2013; 7: 701–7. PubMed Abstract
  59. Potapovich AI, Lulli D, Fidanza P, Kostyuk VA, De Luca C, Pastore S, et al. Plant polyphenols differentially modulate inflammatory responses of human keratinocytes by interfering with activation of transcription factors NFkappaB and AhR and EGFR-ERK pathway. Toxicol Appl Pharmacol 2011; 255: 138–49. PubMed Abstract | Publisher Full Text
  60. Sahni M, Ambrosetti DC, Mansukhani A, Gertner R, Levy D, Basilico C. FGF signaling inhibits chondrocyte proliferation and regulates bone development through the STAT-1 pathway. Genes Dev 1999; 13: 1361–6. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  61. Sahni M, Raz R, Coffin JD, Levy D, Basilico C. STAT1 mediates the increased apoptosis and reduced chondrocyte proliferation in mice overexpressing FGF2. Development 2001; 128: 2119–29. PubMed Abstract
  62. Tian C, Ye X, Zhang R, Long J, Ren W, Ding S, et al. Green tea polyphenols reduced fat deposits in high fat-fed rats via erk1/2-PPARgamma-adiponectin pathway. PLoS One 2013; 8: e53796. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  63. Krejci P, Masri B, Fontaine V, Mekikian PB, Weis M, Prats H, et al. Interaction of fibroblast growth factor and C-natriuretic peptide signaling in regulation of chondrocyte proliferation and extracellular matrix homeostasis. J Cell Sci 2005; 118: 5089–100. PubMed Abstract | Publisher Full Text
  64. Yasoda A, Komatsu Y, Chusho H, Miyazawa T, Ozasa A, Miura M, et al. Overexpression of CNP in chondrocytes rescues achondroplasia through a MAPK-dependent pathway. Nat Med 2004; 10: 80–6. PubMed Abstract | Publisher Full Text
  65. Beenken A, Mohammadi M. The FGF family: biology, pathophysiology and therapy. Nat Rev Drug Discov 2009; 8: 235–53. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  66. Blunt AG, Lawshe A, Cunningham ML, Seto ML, Ornitz DM, MacArthur CA. Overlapping expression and redundant activation of mesenchymal fibroblast growth factor (FGF) receptors by alternatively spliced FGF-8 ligands. J Biol Chem 1997; 272: 3733–8. PubMed Abstract | Publisher Full Text
  67. Valverde-Franco G, Binette JS, Li W, Wang H, Chai S, Laflamme F, et al. Defects in articular cartilage metabolism and early arthritis in fibroblast growth factor receptor 3 deficient mice. Hum Mol Genet 2006; 15: 1783–92. PubMed Abstract | Publisher Full Text
  68. Ellman MB, Yan D, Ahmadinia K, Chen D, An HS, Im HJ. Fibroblast growth factor control of cartilage homeostasis. J Cell Biochem 2013; 114: 735–42. PubMed Abstract | PubMed Central Full Text | Publisher Full Text
  69. Otani Y, Komura M, Komura H, Ishimaru T, Konishi K, Komuro H, et al. Optimal amount of basic fibroblast growth factor in gelatin sponges incorporating beta-tricalcium phosphate with chondrocytes. Tissue Eng Part A 2015; 21: 627–36. PubMed Abstract | Publisher Full Text
  70. Krejci P, Bryja V, Pachernik J, Hampl A, Pogue R, Mekikian P, et al. FGF2 inhibits proliferation and alters the cartilage-like phenotype of RCS cells. Exp Cell Res 2004; 297: 152–64. PubMed Abstract | Publisher Full Text
  71. L’Hote CGM, Knowles MA. Cell responses to FGFR3 signalling: growth, differentiation and apoptosis. Exp Cell Res 2005; 304: 417–31. PubMed Abstract | Publisher Full Text
  72. Joven J, March I, Espinel E, Fernandez-Arroyo S, Rodriguez-Gallego E, Aragones G, et al. Hibiscus sabdariffa extract lowers blood pressure and improves endothelial function. Mol Nutr Food Res 2014; 58: 1374–8. PubMed Abstract | Publisher Full Text
  73. Yamashita A, Morioka M, Kishi H, Kimura T, Yahara Y, Okada M, et al. Statin treatment rescues FGFR3 skeletal dysplasia phenotypes. Nature 2014; 513: 507–11. PubMed Abstract | Publisher Full Text
  74. Yudoh K, Karasawa R. Statin prevents chondrocyte aging and degeneration of articular cartilage in osteoarthritis (OA). Aging 2010; 2: 990–8. PubMed Abstract | PubMed Central Full Text
  75. Masana L, Camprubi M, Sarda P, Sola R, Joven J, Turner PR. The Mediterranean-type diet: is there a need for further modification? Am J Clin Nutr 1991; 53: 886–9. PubMed Abstract