GET THE APP

Pro-angiogenic Activity Assay of Chondroitin Sulfate and Glucosam
Journal of Glycobiology

Journal of Glycobiology
Open Access

ISSN: 2168-958X

+44 1478 350008

Research Article - (2017) Volume 6, Issue 3

Pro-angiogenic Activity Assay of Chondroitin Sulfate and Glucosamine Sulfate on Vascular Network of Mouse and of Chick Embryo Chorioallantoic Membrane

Fernanda Katharine De Souza Lins Borba1,2, Edbhergue Ventura Lola Costa1, Viviane Aparecida Balvedi Polli2, Daniela Sousa Coelho2, Marcelo Maraschin2, Paulo Fernando Dias2 and Romildo Albuquerque Nogueira1*
1Department of Animal Morphology and Physiology, Laboratory of Theoretical, Experimental and Computational Biophysics, Rural Federal University of Pernambuco, Brazil
2Department of Cell Biology, Laboratory of Studies in Bioactivity and Animal Morphogenesis, Embriology and Genetics, Federal University of Santa Catarina, Brazil
*Corresponding Author: Romildo Albuquerque Nogueira, Department of Animal Morphology and Physiology, Rural Federal University of Pernambuco, Dois Irmãos, 52171-900, Recife, Pernambuco, Brazil, Tel: 5581-33206395 Email:

Abstract

Objective: Target of this study was to test the capacity of chondroitin sulfate (CS) and glucosamine sulfate (GS) to induce in vivo angiogenesis. Methods: The proangiogenic activity of these compounds was analyzed through the assays in chorioallantoic membrane (CAM) of chick embryo and dorsal skin vascularization in mice, but before was realized a cell viability assay with human umbilical veins endothelial cells (HUVEC). Results: In the viability assay, concentrations tested between 30 and 3000 μg/ml showed a reduction of viable HUVEC number. In the CAM assay, CS and GS in an amount 2.0 mg/implant increased the vessels number as compared to control (phosphate buffered saline-PBS). In the assay of the dorsal skin vascularization of adult Swiss mice, the groups treated with CS (2 mg/implant; Gelfoam plug) exhibited an increase in the vessels number into plugs (0.52 ± 0.08 g/dl; measured as plug-hemoglobin content), a similar effect to that promoted by Fibroblast growth factor-2 (FGF-2; 50 ng/implant) (0.53 ± 0.1 g/dl). However the group treated with GS did not exhibit significant effect on mice skin vascularization. Conclusion: CS was capable to promote angiogenesis on CAM and dorsal skin vascularization, but GS only had pro-angiogenic activity in CAM vascular network.

<

Keywords: Chondroitin sulphate; Glucosamine sulphate; Angiogenesis; Chorioallantoic membrane; Matrigel plug

Abbreviations

ANOVA: Analysis of Variance; CAM: Chorioallantoic Membrane; CS: Chondroitin Sulphate; CSPG: Chondroitin Sulfate Proteoglycan; DBC: Box-Counting Dimension; DMSO: Dimethyl Sulfoxide; FBS: Fetal Bovine Serum; FGF-2: Fibroblast Growth Factor-2; GAG: Glycosaminoglycan; GS: Glucosamine Sulphate; HS: Heparin Sulphate; HSPGs: Heparin Suphate Proteoglycans; HUVEC: Human Umbilical Veins Endothelial Cells; ICAM-1: Intercellular Adhesion Molecule-1; MTT: 3-(4,5- Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide; NG2: Neuron-glial 2; NRP1: Neuropilin-1; PG: Proteoglycan; PBS: Phosphate Buffered Saline PDGFB: Platelet-Derived Growth Factor Subunit B; RPMI: Roswell Park Memorial Institute; SEM: Standard Error of the Median; VEGF: Vascular Endothelial Growth Factor; VEGFR: Vascular Endothelial Growth Factor Receptor; YSM: Yolk Sac Membrane; TGF-β: Transforming Growth Factor β

Introduction

Angiogenesis is the blood vessels sprouting from the pre-existing vessels, leading vascular network remodeling [1,2]. This remodeling is characterized by luminal diameter expansion of newly formed vessels in response to increased blood flow [3,4]. The formation of new blood vessels involves steps such as proteolytic degradation of extracellular matrix, migration by chemotaxis, adhesion, proliferation and differentiation of endothelial cells; finally emerging a new tubular structure to the bloodstream [5-7]. Addition of the endothelial cells, the mural cells (muscle cells and pericytes) participate in the vessels morphogenesis guided by growth factors interaction with their receptors; furthermore also there is a contribution of other molecules to blood vessels formation [8-10].

Angiogenesis is stimulated by growth factors, including particularly Vascular endothelial growth factor (VEGF) and FGF-2 [11-13]. These growth factors promote several angiogenesis steps, including endothelial cells interaction with extracellular matrix and degradation of this matrix by own endothelial cells [14,15]. This interaction between growth factors with their respective receptors is necessary to promote the angiogenesis. Some polysaccharides can be involved in this interaction, they are known as Glycosaminoglycans (GAGs) [16].

GAGs are unbranched heteropolysaccharides composed of repeating disaccharide units that consist of either sulfated or nonsulfated monosaccharides [17,18]. Disaccharide repeating units are composed uronic acids (D-glucoronic acid or L-iduronic acid) and amino sugar (D-galactosamine or D-glucosamine) [19]. GAGs can differ according to the sulfation, as well as the presence of amino sugars and uronic acids [18]. Non-sulfated GAGs include hyaluronic acid, whereas sulfated GAGs can possess galactosamine in their chains (CS and Dermatan sulfate), glucosamine (heparin and Heparin sulfate- HS) [18] or to contain galactosamine without uronic acids (Keratan sulfate) [18].

GAGs as HS and CS may be attached to a core protein constituting a proteoglycan (PG) that is able to regulate the growth factors activities [16,17,20,21]. HS with its PGs (HSPGs) regulate angiogenesis through capability to bind to growth factors (VEGF and FGF-2) and also to their receptors [16]. Binding of VEGF165 to HS is mediated by amino acid C-terminal heparin binding domain, meanwhile the connection of HS with VEGF receptor 2 (VEGFR2) probably is realized through a ten amino acid sequence between Ig-like domains 6 and 7 of this receptor [22]. The heparin binding domain of FGF-2 is discontinuous and located in the N-terminal and in the C-terminal part, while the heparin binding site to FGF receptor 1 (FGFR1) is contained in the Iglike module II of this receptor [22].

PGs are localized on cellular surface, basement membrane or extracellular matrix [17]. Syndecans (syndecan-1 to 4) and betaglycan are examples of PG (compound of HS and CS) present on cellular membrane [23-25]. Neuropilin-1 (NRP1) also is a membrane PG of endothelial cells, formed for CS or HS [25], NRP1 is VEGFR2 coreceptor [26,27] and is essential to embryonic angiogenesis and vascular development [28]. NRP1 can interact with heparin-biding isoforms VEGF-A, VEGF-B and VEGF-E [29].

Our aim was to test the capacity of CS and GS to induce angiogenesis, utilizing CAM of chick embryos (embryonic angiogenesis) and adult mice skin (advanced angiogenesis).

Materials and Method

Materials

The materials utilized in assay were: CS and GS, both with purity above 90% (Phytomare Company, Governador Celso Ramos, SC, Brazil); dimethyl sulfoxide (DMSO), FGF-2 (F0291), methylcellulose, were purchased from Sigma-Aldrich (St. Louis, MO, USA); MTT (3- (4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) was purchased from Invitrogen (Grand Island, N.Y., USA); FBS, Lglutamine, penicillin/streptomycin, RPMI (Roswell Park Memorial Institute) 1640 medium were purchased from Gibco (Auckland, New Zealand); Gelfoam (Pharmacia and UpJohn, Kalamazoo, Mi, USA).

Cell viability assay

Toxicity test was performed on HUVEC. The HUVEC were seeded (5.0 × 106 cells/well) in RPMI-1640 medium with 2% FBS, 1% antibiotic (penicillin 1000 UI/ml+streptomycin 250 mg/ml) and 1% Lglutamine 200 mM. The cells suspension was distributed in culture plate with 96-well plates. The plates were incubated at 37°C in CO2 incubator. After 24 h of incubation, the cells were treated with increasing concentrations of CS and GS (1-3.000 μg/ml). After 3 days, the medium was removed and wells were washed several times with 100 μl of PBS. Next, the evaluation of viability was carried out according to Carmichael et al. [30] and Dias et al. [31], by colorimetric essay of MTT. The RPMI-1640 medium was added to MTT solution (5 mg/ml in PBS) at a ratio of MTT solution to medium of 1:10. The plates were incubated in a humidified atmosphere containing 5% CO2 at 37°C for 4 h, the medium was aspired and 200 ml DMSO was added to each well to dissolve the formazan crystals. The plates were agitated on a plate shaker for 5 min and the absorbance at 540 nm was determined using a spectra rainbow microplate reader (Tecan, Männedorf, Switzerland). The results are expressed as percent of control (incubation of HUVEC in RPMI-1640 medium alone). All experiments were performed in triplicate.

Animals

All animal studies were carried out in accordance with the procedures outlined in protocol number PP00586/2011/CEUA/UFSC, approved by the Local Committee for Care and Ethical Use of Animals in Research (CEUA/UFSC, Florianopolis, SC, Brazil).

CAM assay

Pathogen-free fertilized chicken eggs (Ross strain, n=6 per experimental group, n=48 in total) were supplied by poultry producers (Tyson S.A., São José, SC, Brazil).

The capacity of CS and GS to stimulate in vivo angiogenesis was determined by the CAM of chick embryo, performed according to the method presented by Dias et al. [31]. The eggs were incubated at 37.5°C and 70% humidity. After 48 h of incubation, a window (10 mm diameter) was opened in the eggshell at a position adjacent to the embryo. The treatment in ovo was performed by implanting diskshaped methylcellulose supports (7.5 μl volume, 3 mm diameter; one disk per embryo) containing only each of the compounds: PBS, CS, GS and FGF-2. The methylcellulose disks were implanted on the outer one-third surface of the CAM of 6 day old embryo (E6), where blood capillaries were still growing. After implanting the disks, the windows were closed with black binding cellophane tape. The eggs stayed in incubation until E8. The concentration of CS and GS administered on CAM vascular network ranged from 0.02 to 2.00 mg/disk. Methylcellulose disks containing FGF-2 (50 ng/disk) and disks containing PBS were used as positive and negative controls, respectively. For each egg, images were captured with a Motic 1000 1.3 MP camera (Motic, Causeway Bay, Hong Kong, China) coupled to a stereomicroscope (20x magnification). The vessels in the region around the limit of disk were quantified by calculating the fractal dimension (Figure 1).

Figure

Figure 1: Image skeletonization of CAM vascular network. Image of CAM vascular network with methylcellulose disk (A) and its respective skeletonized image (B).

Image skeletonization of vascular network

The digital images (1240 × 1024 pixels) of vascular network were manually skeletonized using Microsoft Paint to separate the blood vessels from the rest of the images. Each vessel was traced by a line that was 1 pixel thick, then, the images were binarized, resulting white vessels on black background [32,33].

Fractal analysis

The method used to calculate the fractal dimension of CAM vascular network was box counting dimension (DBC). The obtainment of DBC values was calculated by Benoit 1.3 Fractal Analysis System software (Trusoft, St. Petersburg, FL, USA). The DBC was obtained by covering the skeletonized image of CAM vessels with N (r) boxes containing at least one point of the image and then repeating with boxes of different sizes. The value of the slope of a plot of a double log of N (r) in function on the sides of boxes r [33] is the DBC and can be calculated through the following equation:

where ε is the small variation in the box size.

Gelfoam plug assay

This in vivo assay was based in the method described by Dias et al. [Dias]. Male Swiss mice (Mus musculus), pathogen-free, were acquired from the Central Biotery of Federal University of Santa Catarina (Florianopolis, Brazil). The male Swiss mice of 8 weeks old with an average body weight of 27 ± 2 g were housed in a light-controlled room (lit from 7:00 A.M. to 7:00 P.M.) with temperature of 24 ± 1°C and fed on sterilized animal feed and water ad libitum.

The groups were treated with CS, GS (2 mg/animal) and FGF-2 (50 ng/animal). PBS (vehicle) and FGF-2 were used, respectively, as negative and positive controls. Each compound was adsorbed (50 μl) in a sterile compressed sponge (Gelfoam plug; 6 mm diameter × 3 mm). Gelfoam plug was implanted subcutaneously into the rear right flank of a mouse (n=6). The mice were killed 2 weeks later by CO2 inhalation and the skin was carefully pulled away to expose the intact Gelfoam plug. The amount of hemoglobin inside the Gelfoam plug was measured using Drabkin reagent as a quantifiable index of blood vessel formation [34]. The hemoglobin concentration is expressed as milligrams per deciliter. The calculation is based on a hemoglobin standard measured simultaneously using the following equation described by Lee et al. [35]: sample absorbance/standard absorbance × 10.

Statistical analysis

Experimental data of each assay were evaluated by ANOVA and Tukey´s Post Hoc test. Effect were considered to be statistically significant at p<0.05. Experimental data were summarized and expressed as mean ± SEM.

Results

Effects of CS and GS on HUVEC viability

We evaluated the effect of increased concentrations of CS and GS (1-3.000 μg/ml) on HUVEC lineage cells viability by the MTT toxicity assay. After 48 h treatment, the MTT assay showed that the HUVEC viability was not statistically significant to the treatments between 1-10 μg/ml to both CS and GS (Figure 2). When the treatment was increased from 30 to 3000 μg/ml, was observed that there was reduction of HUVEC viability. In concentration of 30 μg/ml, the CS and GS reduced the amount of viable cells to 65.5 ± 1.25% and 60.3 ± 2.41%, respectively when compared to control group (100%), to pFigure 2 shows the effect for administration of CS and GS in all tested concentrations.

Figure

Figure 2: Effects of CS and GS (1-3000 μg/ml) on HUVEC line cells viability (5.0 × 106 cells/well). After 48 h of exposition to the test compounds, the percentage of viable cells was determined by incubation (4 h) with MTT. The experiments were done in triplicate, and the results are expressed as percentage of the control group (medium: RPMI-1640). Each column with respective vertical bar represents mean ± SEM, the columns with asterisk denote statistically significant differences with p<0.05, in relation to the control group (ANOVA followed by Tukey test).

CAM assay

CAM assay was used to determine whether CS and GS displayed pro-angiogenenic activity. We evaluated the vascularization around the methylcellulose disk in chick embryo CAM (implanted at E6). Figure 3 show the results of fractal dimensions DBC for the groups treated with CS and GS, respectively, at E8.

Figure

Figure 3: Stimulatory effect to concentrations of 0, 2-2 mg/ml of both chondroitin sulphate (A) and glucosamine sulfate (B) on vascularization of the 8 day YSM. Results are quantified by boxcounting fractal dimension (DBC). The controls were performed with vehicle (PBS, negative control) and FGF-2 (50 ng/disk, positive control). Each column with respective vertical bar represents the mean ± SEM of six eggs. *p<0.05, versus negative control (ANOVA and Tukey as a post hoc test).

Figure 3A and 3B display a significant increase (p<0.05) in the density of blood vessels to the highest tested concentration (2 mg/disk) of CS (DBC=1.30 ± 0.03) and GS (DBC=1.28 ± 0.04) when compared to the negative control group (DBC=1.19 ± 0.03). In addition, this concentration of CS and GS had a similar effect to the FGF-2 (DBC=1.34 ± 0.03) in promoting neovascularization. However, significant difference (p<0.05) was no observed for the fractal dimensions at concentrations of CS and GS below 2 mg/disk, compared to the negative control group (PBS).

Gelfoam plug assay

We conducted an assay in dorsal subcutaneous vascularization of mice with 8 weeks in order to test whether the pro-angiogenic effects of CS and GS, previously observed in embryonic stage, would also be observed in adult individuals (advanced angiogenesis). In this experiment, we evaluated the formation of blood vessels based on the amount of hemoglobin in the Gelfoam plug implanted under the animal skin.

At the 15th experiment day, negative control group (PBS-treated Gelfoam plugs) clearly showed new blood vessels, with hemoglobin content in the plug of 0.20 ± 0.07 mg/dl. However, Gelfoam plugs containing FGF-2 (positive control) displayed a higher hemoglobin content (0.53 ± 0.1 mg/dl) than negative control (p<0.01, Figure 4). Group treated with CS (2 mg/implant) displayed a higher vessels growth (hemoglobin content of 0.52 ± 0.08 mg/dl) than negative control group (p<0.01). While GS did not induce a significant vessels growth (0.37 ± 0.09 mg/dl) compared to the negative control group.

Figure

Figure 4: Effects of CS and GS (2 mg/animal), as well as combined with FGF-2 (50 ng/animal) in subcutaneously implanted Gelfoam plugs in adult mice, based on hemoglobin content in the plug. The test implants were adsorbed from 50 μl of FGF-2 (50 ng), with or without CS or GS. The negative and positive controls consisted of, respectively, 50 μl of PBS (vehicle) and FGF-2 (50 ng). Each column with respective vertical bar represents the mean ± SEM for at least 5 animals, and the asterisks denote statistically significant differences for **P<0.01.

Discussion

Glucosamine is an aminomonosaccharide which constitutes some GAGs which form the matrix of all connective tissues including articular cartilage, while CS is a GAG linked sulfate group and also composes the articular cartilage [36]. Some studies have reported pro [21,32,37] and anti-angiogenic effects [38-40] of CS, well as proangiogenic effect of GS [32,41,42]. Here, we show that concentrations of CS and GS in the range of 30-3000 μg/ml promoted toxicity to HUVEC (5.0 × 106 cells). GS had a higher toxic effect on cells than CS.

A concentration of 2000 μg/disk of CS and GS was capable to stimulate neovascularization in the CAM of chick embryo, while lower concentrations of these molecules did not have significant effect (Figure 3). In Gelfoam plug implanted under mice skin, CS showed a significant effect on the amount of erythrocytes in relation to control group. Nevertheless, the GS did not show significant effect to Gelfoam plug assay. According to in vivo assays, there is a threshold concentration of CS and GS to perform angiogenic activity, well as a high concentration can result in toxic effects to cells as shown by in vitro assay.

In relation to the angiogenic activity, CS provoked a higher effect on vascular density in relation to the GS, as shown in the CAM and Gelfoam plug assay. We have observed in our previous work that CS and GS were capable stimulate vasculogenesis and angiogenesis in the yolk sac membrane (YSM) of chick embryo through fractal analysis [32]. However 1 μg/ml of GS produced a similar increase in the vascular network obtained by a dose of 100 μg/ml of CS [32], thus contradicting the results shown in the current paper in which the angiogenic activity of CS was higher, according to the fractal dimension and amount of hemoglobin obtained in Gelfoam plug assay. GS had a more effective action on vasculogenesis and early angiogenesis (YSM assay and CAM) than later angiogenesis (Gelfoam plug assay). The availability of GS at the emergence and initial growing of vessels seems to contribute more to expansion of the vascular network than at later angiogenesis. Since GS can participate in the HSPGs synthesis by vascular endothelium, thus contributing to vessels development [22].

On the other hand, CS presented a higher action on mature angiogenesis. The CS is a heteropolysaccharide which can bind to proangiogenic factors such as VEGF-A, TGF-β (Transforming growth factor β), PDGFB (Platelet-derived growth factor subunit B) [21]. The CS can act on signaling of TGF-β, this growth factor is able to maintain the endothelial cells quiescence, induces vessels maturation and influences expression and angiogenic factors activities like VEGF [21,43]. CS is capable to regulate growth factors-mediated cell migration implicating in tumor angiogenesis [24]. The study realized by Le Jan et al. [21] also indicates the involvement of CS in angiogenesis sprouting. CSPGs are able to modulate several steps of angiogenesis [21]. There is a CSPG known as Neuron-glial 2 (NG2) that is expressed on the surface of pericytes during vasculogenesis and angiogenesis [44]. This CSPG is considerable element in promoting endothelial cells migration and morphogenesis in the early stages of neovascularization [45]. Also Tapon-Bretaudière et al. [37] reported an increase of vascular tubes formation by endothelial cells in the presence of FGF-2 in HUVECs treated with the fucosylated CS obtained from sea cucumber, which has similar chemical structure to mammalian CS.

Conclusion

In our study, concentrations between 30 and 3000 μg/ml were enough to decrease the HUVEC viability. Fractal analysis of vascular network of chick embryo chorioallantoic membrane revealed the proangiogenic effect of chondroitin and glucosamine sulfates (2 mg/disk). Also the evaluation of hemoglobin content in the Gelfoam plug implanted under the mouse skin permitted to identify angiogenic capacity of chondroitin sulfate (2 mg/disk). Nevertheless the Gelfoam plug assay showed that glucosamine sulfate did not have significant angiogenic effect.

Acknowledgement

We thank Dr. Maria Beatriz da Rocha Veleirinho (Plant Morphogenesis and Biochemistry Laboratory/CCA,-UFSC) by the valuable collaboration in the development at this work. We also thank Giovanni Loss and Lisiê Silva by their technical assistance. We are grateful to the Research support center of UFRPE (CENAPESQ) for technical support.

This work was funded by Brazilian support agencies: Coordination for the Improvement of Higher Education Personnel (CAPES), Foundation for Science and Technology Support in Pernambuco (FACEPE), National Council for Scientific and Technological Development (CNPq).

References

  1. Ribatti D, Crivellato E (2012) Sprouting angiogenesis, a reappraisal. Dev Biol 372: 157-165.
  2. Logsdon EA, Finley SD, Popel AS, Gabhann FM (2014) A systems biology view of blood vessel growth and remodeling. J Cell Mol Med 18: 1491-1508.
  3. Emmett MS, Dewing D, Pritchard-Jones RO (2011) Angiogenesis and melanoma-from basic science to clinical trials. Am J Cancer Res 1: 852-868.
  4. Kubota Y (2012) Tumor angiogenesis and anti-angiogenic therapy. The Keio J Med 61: 47-56.
  5. Ding Z, Lambrechts A, Parepally M, Roy P (2006) Silencing profilin-1 inhibits endothelial cell proliferation, migration and cord morphogenesis. J Cell Sci 119: 4127-4137.
  6. Katoh M (2013) Therapeutics targeting angiogenesis: Genetics and epigenetics, extracellular miRNAs and signaling networks. Int J Mol Med 32: 763-767.
  7. Ambler CA, Schmunk GM, Bautch VL (2003) Stem cell-derived endothelial cells/progenitors migrate and pattern in the embryo using the VEGF signaling pathway. Dev Biol 257: 205-219.
  8. Fujimoto A, Onodera H, Mori A, Isobe N, Yasuda S, et al. (2004) Vascular endothelial growth factor reduces mural cell coverage of endothelial cells and induces sprouting rather than luminal division in an HT1080 tumour angiogenesis model. Int J Clin Exp Pathol 85: 355-364.
  9. Herbert SP, Stainier DYR (2011) Molecular control of endothelial cell behaviour during blood vessel morphogenesis. Nat Rev Mol Cell Biol 12: 551-564.
  10. Yun YR, Won JE, Jeon E, Lee S, Kang W, et al. (2010) Fibroblast growth factors: biology, function, and application for tissue regeneration. J Tissue Eng 1: 1-18.
  11. Madanecki P, Kapoor N, Bebok Z, Ochocka R, Collawn JF, et al. (2013) Regulation of angiogenesis by hypoxia: The role of microRNA. Cell Mol Biol Lett 18: 47-57.
  12. Maruotti N, Annese T, Cantatore FP, Ribatti D (2013) Macrophages and angiogenesis in rheumatic diseases. Vasc Cell 5: 1-8.
  13. Campbell NE, Kellenberger L, Greenaway J, Moorehead RA, Linnerth- Petrik NM, et al. (2010) Extracellular matrix proteins and tumor angiogenesis. J Oncol 1: 1-13.
  14. Campbell NE, Kellenberger L, Greenaway J, Moorehead RA, Linnerth-Petrik NM, et al. (2010) Extracellular matrix proteins and tumor angiogenesis. J Oncol 1: 1-13.
  15. Eming SA, Hubbell JA (2011) Extracellular matrix in angiogenesis: dynamic structures with translational potential. Exp Dermatol 20: 600-613.
  16. Chiodelli P, Bugatti A, Urbinati C, Rusnati M (2015) Heparin/heparan sulfate proteoglycans glycomic interactome in angiogenesis: Biological implications and therapeutical use. Molecules 20: 6342-6388.
  17. Yip GW, Smollich M, Götte M (2006) Therapeutic value of glycosaminoglycans in cancer. Mol Cancer Ther 5: 2139-2148.
  18. Afratis N, Gialeli C, Nikitovic D, Tsegenidis T, Karousou E, et al. (2012) Glycosaminoglycans: Key players in cancer cell biology and treatment. Febs J 279: 1177-1197.
  19. Gandhi NS, Mancera RL (2008) The structure of glycosaminoglycans and their interactions with proteins. Chem Biol Drug Des 72: 455-482.
  20. Coombe DR (2008) Biological implications of lycosaminoglycan interactions with haemopoietic cytokines. Immunol Cell Biol 86: 598-607.
  21. Le Jan S, Hayashi M, Kasza Z, Eriksson I, Bishop JR, et al. (2012) Functional Overlap Between Chondroitin and Heparan Sulfate Proteoglycans During VEGF- Induced Sprouting Angiogenesis. Arterioscler.Thromb Vasc Biol 32: 1255-1263.
  22. van Wijk, XMR, van Kuppevelt TH (2014) Heparan sulfate in angiogenesis: A target for therapy. Angiogenesis 17: 443-462.
  23. Deepa SS, Yamada S, Zako M, Goldberger O, Sugahara K (2004) Chondroitin sulfate chains on Syndecan-1 and Syndecan-4 from normal murine mammary gland epithelial cells are structurally and functionally distinct and cooperate with heparan sulfate chains to bind growth factors. J Biol Chem 279: 37368-37376.
  24. Pantazaka E, Papadimitriou E (2014) Chondroitin sulfate-cell membrane effectors as regulators of growth factor-mediated vascular and cancer cell migration. Biochim Biophys Acta 1840: 2643-2650.
  25. Sarrazin S, Lamanna WC, Esko JD (2011) Heparan sulfate proteoglycans. Cold Spring Harb Perspect Biol 3: a004952.
  26. Gelfand MV, Hagan N, Tata A, Oh WJ, Lacoste B, et al. (2014) Neuropilin-1 functions as a VEGFR2 co-receptor to guide developmental angiogenesis independent of ligand binding. Elife 3: e03720.
  27. Evans IM, Yamaji M, Britton V, Pellet-Many C, Lockie C, et al. (2011) Neuropilin-1 signaling through p130Cas tyrosine phosphorylation is essential for growth factor-dependent migration of glioma and endothelial cells. Mol Cell Bio 31: 1174-1185.
  28. Plein A, Fantin A, Ruhrberg C (2014) Neuropilin regulation of angiogenesis, arteriogenesis, and vascular permeability. Microcirculation 21: 315-323.
  29. Evans IM, Yamaji M, Britton V, Pellet-Many C, Lockie C, et al. (2011) Neuropilin-1 signaling through p130Cas tyrosine phosphorylation is essential for growth factor-dependent migration of glioma and endothelial cells. Mol Cell Bio 31: 1174-1185.
  30. Djordjevic S, Driscoll PC (2013) Targeting VEGF signalling via the neuropilin co-receptor. Drug Discov Today 18: 447-455.
  31. Carmichael J, Degraff WG, Gazdar AF, Minn JD, Mitchell JB (1987) Evaluation of a tetrazolium-based semiautomated colorimetric assay: Assessment of chemosensitivity testing. Cancer Res 47: 936-942.
  32. Dias PF, Siqueira JM, Vendruscolo LF, Neiva TJ, Gagliardi AR, et al. (2005) Antiangiogenic and antitumoral properties of a polysaccharide isolated from the seaweed Sargassum stenophyllum. Cancer Chemother Pharmacol 56: 436-446.
  33. Borba FKSL, Felix GLQ, Costa EVL, Silva L, Dias PF, et al. (2016) Fractal analysis of extra-embryonic vessels of chick embryos under the effect of glucosamine and chondroitin sulfates. Microvasc Res 105: 114-118.
  34. Costa EVL, Jimenez GC, Barbosa CTF, Nogueira RA (2013) Fractal analysis of extra-embryonic vascularization in japanese quail embryos exposed to extremely low frequency magnetic fields. Bioelectromagnetics 34: 114 -121.
  35. Drabkin DL, Austin JH (1932) Spectrophotometric constants for common hemoglobin derivatives in human, dog, and rabbit blood. J Biol Chem 98: 719-722.
  36. Lee YS, Yang HO, Shin KH, Choi HS, Jung SH, et al. (2003) Suppression of tumor growth by a new glycosaminoglycan isolated from the African giant snail Achatina fulica. Eur J Pharmacol 465: 191-198.
  37. Hathcock JN, Shao A (2007) Risk assessment for glucosamine and chondroitin sulfate. Regul Toxicol Pharmacol 47: 78-83.
  38. Tapon-Bretaudière J, Chabut D, Zierer M, Matou S, Helley D, et al. (2002) A fucosylated chondroitin sulfate from echinoderm modulates in vitro fibroblast growth factor 2-dependent angiogenesis. Mol Cancer Res 1: 96-102.
  39. Wang J, Svendsen A, Kmiecik J, Immervoll H, Skaftnesmo OK, et al. (2011) Targeting the NG2/CSPG4 proteoglycan retards tumour growth and angiogenesis in preclinical models of gbm and melanoma. Plos One 6: 1-14.
  40. Calamia V, Lourido L, Fernández-Puente P, Mateos J, Rocha B, et al. (2012) Secretome analysis of chondroitin sulfate-treated chondrocytes reveals anti-angiogenic, anti-inflammatory and anti-catabolic properties. Arthritis Res Ther 14: 1-12.
  41. Scherer SS, Pietramaggiori G, Matthews JC, Gennaoui A, Demcheva M, et al. (2011) Poly-N-Acetyl glucosamine fibers induce angiogenesis in ADP Inhibitor-treated diabetic mice. J Trauma 71: S183-S186.
  42. Lambert C, Mathy-Hartert M, Dubuc JE, Montell E, Vergés J, et al. (2012) Characterization of synovial angiogenesis in osteoarthritis patients and its modulation by chondroitin sulfate. Arthritis Res Ther 14: 1-11.
  43. Vournakis JN, Eldridge J, Demcheva M, Muise-Helmericks RC (2008) Poly-n-acetyl glucosamine nanofibers regulate endothelial cell movement and angiogenesis: Dependency on integrin activation of Ets1. J Vasc Res 45: 222-232.
  44. Ma J, Wang Q, Fei T, Han JDJ, Chen YG (2007) MCP-1 mediates TGF-β-induced angiogenesis by stimulating vascular smooth muscle cell migration. Blood 109: 987-994.
Citation: Borba FKSL, Costa EVL, Polli VAB, Coelho DS, Maraschin M, et al. (2017) Pro-angiogenic Activity Assay of Chondroitin Sulfate and Glucosamine Sulfate on Vascular Network of Mouse and of Chick Embryo Chorioallantoic Membrane. J Glycobiol 6: 129.

Copyright: © 2017 Borba FKSL, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top