Fish Scale Collagen Peptides Protect against CoCl2/TNF- α-Induced Cytotoxicity and Inflammation via Inhibition of ROS, MAPK, and NF- κ B Pathways in HaCaT Cells

Fazli Subhan, Hae Yeong Kang, Yeseon Lim, Muhammad Ikram, Sun-Yong Baek, Songwan Jin, Young Hun Jeong, Jong Young Kwak, Sik Yoon, Fazli Subhan, Hae Yeong Kang, Yeseon Lim, Muhammad Ikram, Sun-Yong Baek, Songwan Jin, Young Hun Jeong, Jong Young Kwak, Sik Yoon

Abstract

Skin diseases associated with inflammation or oxidative stress represent the most common problem in dermatology. The present study demonstrates that fish scale collagen peptides (FSCP) protect against CoCl2-induced cytotoxicity and TNF-α-induced inflammatory responses in human HaCaT keratinocyte cells. Our study is the first to report that FSCP increase cell viability and ameliorate oxidative injury in HaCaT cells through mechanisms mediated by the downregulation of key proinflammatory cytokines, namely, TNF-α, IL-1β, IL-8, and iNOS. FSCP also prevent cell apoptosis by repressing Bax expression, caspase-3 activity, and cytochrome c release and by upregulating Bcl-2 protein levels in CoCl2- or TNF-α-stimulated HaCaT cells. In addition, the inhibitory effects of FSCP on cytotoxicity and the induction of proinflammatory cytokine expression were found to be associated with suppression of the ROS, MAPK (p38/MAPK, ERK, and JNK), and NF-κB signaling pathways. Taken together, our data suggest that FSCP are useful as immunomodulatory agents in inflammatory or immune-mediated skin diseases. Furthermore, our results provide new insights into the potential therapeutic use of FSCP in the prevention and treatment of various oxidative- or inflammatory stress-related inflammation and injuries.

Figures

Figure 1
Figure 1
Stimulatory effects of FSCP on proliferation and viability of HaCaT cells. Treatment of HaCaT cells with FSCP for 24 h significantly enhanced cell proliferation (a) and attenuated the CoCl2-induced (500 μM CoCl2) decrease in cell viability (b). Results are presented as the means ± SD of three independent experiments. #p < 0.05 versus the control and ∗∗p < 0.01 versus the CoCl2-treated group.
Figure 2
Figure 2
Inhibitory effects of FSCP on CoCl2/TNF-α-induced ROS generation in HaCaT cells. Intracellular ROS levels were determined via fluorescence microscopy (a, c) or flow cytometry (b, d) using DCFH-DA. The increased ROS levels induced by 100 μM CoCl2 treatment was significantly attenuated by treatment with 1 mg/mL FSCP for 24 h or NAC for 2 h (a, b). Elevated ROS levels were induced by treatment with 20 ng/mL TNF-α but were reversed by treatment with 1 mg/mL FSCP for 12 or 24 h (c, d). Staining intensities were measured using ImageJ software (a, c). Blue histograms represent ROS negative cells, red histograms represent ROS positive cells, and the median fluorescence intensity (MFI) of ROS expression is plotted along the y-axes (b, d). Results are presented as the means ± SD of three independent experiments. #p < 0.05 versus the control and ∗∗p < 0.01 and ∗∗∗p < 0.001 versus the CoCl2/TNF-α-treated group.
Figure 3
Figure 3
Inhibitory effects of FSCP on CoCl2/TNF-α-induced proinflammatory cytokine expression. The expression of the key proinflammatory cytokines, namely, TNF-α, IL-1β, IL-8, and iNOS, was analyzed via RT-PCR. Elevated levels of TNF-α and iNOS mRNA in CoCl2-treated HaCaT cells were attenuated by treatment with FSCP for 24 h (a, b). Upregulation of IL-1β and IL-8 mRNA in TNF-α-treated HaCaT cells was decreased by treatment with FSCP for 24 h (c, d). Results are presented as the means ± SD of three independent experiments. #p < 0.05 versus the control and ∗p < 0.05, ∗∗p < 0.01, and ∗∗∗p < 0.001 versus the CoCl2/TNF-α-treated group.
Figure 4
Figure 4
Effects of FSCP on CoCl2-induced altered expression of apoptosis-related proteins. The expression of apoptosis-related proteins was analyzed by western blotting. Bcl-2 downregulation and Bax upregulation in CoCl2-treated HaCaT cells were reversed by treatment with FSCP and NAC (a, b). Potentiated levels of caspase-3 activity and cytochrome c release in CoCl2-treated HaCaT cells were attenuated by treatment with FSCP and NAC (c). Results are presented as the means ± SD of three independent experiments. Cl-caspase-3: cleaved-caspase-3. #p < 0.05 versus the control and ∗∗p < 0.01 and ∗∗∗p < 0.001 versus the CoCl2-treated group.
Figure 5
Figure 5
Inhibitory effects of FSCP on CoCl2-induced activation of p38/MAPK signaling pathway. P-p38/MAPK levels were increased in CoCl2-treated HaCaT cells (a). Treatment of HaCaT cells with FSCP and NAC (a) and SB203580 (SB) (b) blocked CoCl2-induced phosphorylation of p38/MAPK. Treatment of HaCaT cells with FSCP and SB203580 (SB) blocked CoCl2-induced intracellular ROS activity (c), and staining intensities were measured using ImageJ software (d). Treatment with FSCP blocked CoCl2-induced cytotoxicity in HaCaT cells (e). Western blot analysis showed that the treatment of HaCaT cells with FSCP and SB203580 (SB) suppressed CoCl2-induced elevated expression of iNOS and Bax (f). Results are presented as the means ± SD of three independent experiments. #p < 0.05 versus the control and ∗p < 0.05, ∗∗p < 0.01, and ∗∗∗p < 0.001 versus the CoCl2-treated group.
Figure 6
Figure 6
Inhibitory effects of FSCP on TNF-α-induced activation of the MAPK signaling pathway. Treatment of HaCaT cells with FSCP did not significantly inhibit TNF-α-induced increase in p38/MAPK phosphorylation (a). Treatment of HaCaT cells with FSCP suppressed TNF-α-induced ERK (b) and JNK (c) activation. Results are presented as the means ± SD of three independent experiments. #p < 0.05 versus the control and ∗∗p < 0.01 versus the TNF-α-treated group.
Figure 7
Figure 7
Inhibitory effects of FSCP on TNF-α-induced activation of the NF-κB signaling pathway. Western blot analysis showed NF-κB p65 levels were decreased in the cytoplasm and increased in the nucleus in TNF-α-treated HaCaT cells (a). Immunofluorescence microscopic assay revealed that FSCP prevented the translocation of NF-κB p65 to the nucleus in TNF-α-treated HaCaT cells (b). Results are presented as the means ± SD of three independent experiments. CE: cytoplasmic extracts; NE: nuclear extract. #p < 0.05 versus the control and ∗∗p < 0.01 and ∗∗∗p < 0.001 versus the TNF-α-treated group.
Figure 8
Figure 8
Schematic representation of the regulatory mechanism of FSCP on CoCl2-/TNF-α-induced oxidative and inflammatory stress in HaCaT cells.

References

    1. Coman G., Blickenstaff N. R., Maibach H. I. Skin and the environment. Reviews on Environmental Health. 2014;29(3):p. 143. doi: 10.1515/reveh-2014-0068.
    1. Bos J. D., Kapsenberg M. L. The skin immune system: progress in cutaneous biology. Immunology Today. 1993;14(2):75–78. doi: 10.1016/0167-5699(93)90062-P.
    1. Kim J. H., Jin Z. W., Murakami G., Cho B. H. Characterization of mesenchymal cells beneath cornification of the fetal epithelium and epidermis at the face: an immunohistochemical study using human fetal specimens. Anatomy and Cell Biology. 2016;49(1):50–60. doi: 10.5115/acb.2016.49.1.50.
    1. Bickers D. R., Athar M. Oxidative stress in the pathogenesis of skin disease. The Journal of Investigative Dermatology. 2006;126(12):2565–2575. doi: 10.1038/sj.jid.5700340.
    1. Narendhirakannan R. T., Hannah M. A. Oxidative stress and skin cancer: an overview. Indian Journal of Clinical Biochemistry. 2013;28(2):110–115. doi: 10.1007/s12291-012-0278-8.
    1. Kim M. Y., Lim Y. Y., Kim H. M., Park Y. M., Kang H., Kim B. J. Synergistic inhibition of tumor necrosis factor-alpha-stimulated pro-inflammatory cytokine expression in HaCaT cells by a combination of rapamycin and mycophenolic acid. Annals of Dermatology. 2015;27(1):32–39. doi: 10.5021/ad.2015.27.1.32.
    1. Kobielak A., Boddupally K. Junctions and inflammation in the skin. Cell Communication & Adhesion. 2014;21(3):141–147. doi: 10.3109/15419061.2014.905930.
    1. Choi Y. H., Yan G. H., Chai O. H., Song C. H. Inhibitory effects of curcumin on passive cutaneous anaphylactoid response and compound 48/80-induced mast cell activation. Anatomy and Cell Biology. 2010;43(1):36–43. doi: 10.5115/acb.2010.43.1.36.
    1. Kumari S., Pasparakis M. Epithelial cell death and inflammation in skin. Current Topics in Microbiology and Immunology. 2015;2015:1–17. doi: 10.1007/82_2015_466.
    1. Mustoe T. A., O’Shaughnessy K., Kloeters O. Chronic wound pathogenesis and current treatment strategies: a unifying hypothesis. Plastic and Reconstructive Surgery. 2006;117(7):35S–41S. doi: 10.1097/01.prs.0000225431.63010.1b.
    1. Yang C., Ling H., Zhang M., et al. Oxidative stress mediates chemical hypoxia-induced injury and inflammation by activating NF-κb-COX-2 pathway in HaCaT cells. Molecules and Cells. 2011;6(6):531–538. doi: 10.1007/s10059-011-1025-3.
    1. Watanabe K., Kawamori T., Nakatsugi S., Wakabayashi K. COX-2 and iNOS, good targets for chemoprevention of colon cancer. Biofactors. 2002;12(1–4):129–133. doi: 10.1002/biof.5520120120.
    1. Wang J. K., Xiong G. M., Luo B., et al. Surface modification of PVDF using non-mammalian sources of collagen for enhancement of endothelial cell functionality. Journal of Materials Science. Materials in Medicine. 2016;27(3):p. 45. doi: 10.1007/s10856-015-5651-8.
    1. Liu C., Xue Y., Sun J. Hydrolyzed fish collagen inhibits inflammatory cytokines secretion in lipopolysaccharide-induced HUVECs. Advances in Materials Research. 2014;1025-1026:570–573. doi: 10.4028/.
    1. Yamamoto K., Igawa K., Sugimoto K., et al. Biological safety of fish (tilapia) collagen. BioMed Research International. 2014;2014:9. doi: 10.1155/2014/630757.630757
    1. Wang L., An X. X., Yang F. M., Xin Z. H., Zhao L. Y., Hu Q. H. Isolation and characterisation of collagens from the skin, scale and bone of deep-sea redfish (Sebastes mentella) Food Chemistry. 2008;108(2):616–623. doi: 10.1016/j.foodchem.2007.11.017.
    1. Cheung R. C., Ng T. B., Wong J. H. Marine peptides: bioactivities and applications. Marine Drugs. 2015;13(7):4006–4043. doi: 10.3390/md13074006.
    1. Zhang F., Wang Z., Xu S. Macroporous resin purification of grass carp fish (Ctenopharyngodon idella) scale peptides with in vitro angiotensin-I converting enzyme (ACE) inhibitory ability. Food Chemistry. 2009;117(3):387–392. doi: 10.1016/j.foodchem.2009.04.015.
    1. Asserin J., Lati E., Shioya T., Prawitt J. The effect of oral collagen peptide supplementation on skin moisture and the dermal collagen network. Journal of Cosmetic Dermatology. 2015;14(4):291–301. doi: 10.1111/jocd.12174.
    1. Ohara H., Iida H., Ito K., Takeuchi Y., Nomura Y. Effects of pro-Hyp, a collagen hydrolysate-derived peptide, on hyaluronic acid synthesis using in vitro cultured synovium cells and oral ingestion of collagen hydrolysates in a guinea pig model of osteoarthritis. Bioscience, Biotechnology, and Biochemistry. 2010;74(10):2096–2099. doi: 10.1271/bbb.100193.
    1. Zhang Z., Wang J., Ding Y., Dai X., Li Y. Oral administration of marine collagen peptides from chum Salmon skin enhances cutaneous wound healing and angiogenesis in rats. Journal of the Science of Food and Agriculture. 2011;91(12):2173–2179. doi: 10.1002/jsfa.4435.
    1. Liu C., Sun J. Potential application of hydrolyzed fish collagen for inducing the multidirectional differentiation of rat bone marrow mesenchymal stem cells. Biomacromolecules. 2015;15(1):436–443. doi: 10.1021/bm401780v.
    1. Ding C. H., Li Q., Xiong Z. Y., Zhou A. W., Jones G., Xu S. Y. Oral administration of type II collagen suppresses pro-inflammatory mediator production by synoviocytes in rats with adjuvant arthritis. Clinical and Experimental Immunology. 2003;132(3):416–423. doi: 10.1046/j.1365-2249.2003.02167.x.
    1. Zou W., Yan M., Xu W., et al. Cobalt chloride induces PC12 cells apoptosis through reactive oxygen species and accompanied by AP-1 activation. Journal of Neuroscience Research. 2001;64(6):646–653. doi: 10.1002/jnr.1118.
    1. Schmalz G., Schweikl H., Hiller K. A. Release of prostaglandin E2, IL-6 and IL-8 from human oral epithelial culture models after exposure to compounds of dental materials. European Journal of Oral Sciences. 2000;108(5):442–448. doi: 10.1034/j.1600-0722.2000.108005442.x.
    1. Okayama Y. Oxidative stress in allergic and inflammatory skin diseases. Current Drug Targets-Inflammation & Allergy. 2005;4(4):517–519. doi: 10.2174/1568010054526386.
    1. Aleman A., Martinez-Alvarez O. Marine collagen as a source of bioactive molecules. A Review. Natural Products Journal. 2013;3(2):105–114. doi: 10.2174/2210315511303020005.
    1. Mendis E., Rajapakse N., Byun H. G., Kim S. K. Investigation of jumbo squid (Dosidicus gigas) skin gelatin peptides for their in vitro antioxidant effects. Life Sciences. 2005;77(17):2166–2178. doi: 10.1016/j.lfs.2005.03.016.
    1. Kim S. K., Kim Y. T., Byun H. G., Nam K. S., Joo D. S., Shahidi F. Isolation and characterization of antioxidative peptides from gelatin hydrolysate of Alaska. Journal of Agricultural and Food Chemistry. 2001;49(4):1984–1989. doi: 10.1021/jf000494j.
    1. Ngo D. H., Ryu B., Vo T. S., Himaya S. W. A., Wijesekara I., Kim S. K. Free radical scavenging and angiotensin-I converting enzyme inhibitory peptides from Pacific cod (Gadus macrocephalus) skin gelatin. International Journal of Biological Macromolecules. International Journal of Biological Macromolecules. 2011;49(5):1110–1116. doi: 10.1016/j.ijbiomac.2011.09.009.
    1. Himaya S. W. A., Ngo D. H., Ryu B., Kim S. K. An active peptide purified from gastrointestinal enzyme hydrolysate of Pacific cod skin gelatine attenuates angiotensin-1 converting enzyme (ACE)activity and cellular oxidative stress. Food Chemistry. 2012;132(4):1872–1882. doi: 10.1016/j.foodchem.2011.12.020.
    1. Mohanty D. P., Mohapatra S., Misra S., Sahu P. S. Milk derived bioactive peptides and their impact on human health - a review. Saudi Journal of Biological Sciences. 2016;23(5):577–583. doi: 10.1016/j.sjbs.2015.06.005.
    1. Ito N., Fukushima S., Hasegawa A., Shibata M., Ogiso T. Carcinogenicity of butylated hydroxyanisole in F344 rats. Journal of the National Cancer Institute. 1983;70(2):343–347. doi: 10.1093/jnci/70.2.343.
    1. Yadav N., Dwivedi A., Mujtaba S. F., et al. Photosensitized mefloquine induces ROS-mediated DNA damage and apoptosis in keratinocytes under ambient UVB and sunlight exposure. Cell Biology and Toxicology. 2014;30(5):253–268. doi: 10.1007/s10565-014-9280-7.
    1. Jeon Y. J., Song K. S., Han H. J., Park S. H., Chang W., Lee M. Y. Rosmarinic acid inhibits chemical hypoxia-induced cytotoxicity in primary cultured rat hepatocytes. Archives of Pharmacal Research. 2014;37(7):907–915. doi: 10.1007/s12272-013-0234-z.
    1. Yang C., Ling H., Zhang M., et al. Oxidative stress mediates chemical hypoxia-induced injury and inflammation by activating NF-κb-COX-2 pathway in HaCaT cells. Molecules and Cells. 2011;31(6):531–538. doi: 10.1007/s10059-011-1025-3.
    1. Wang J., Xu M., Liang R., Zhao M., Zhang Z., Li Y. Oral administration of marine collagen peptides prepared from chum salmon (Oncorhynchus keta) improves wound healing following cesarean section in rats. Food & Nutrition Research. 2015;59:p. 26411. doi: 10.3402/fnr.v59.26411.
    1. Xu L., Dong W., Zhao J., Xu Y. Effect of marine collagen peptides on physiological and neurobehavioral development of male rats with perinatal asphyxia. Marine Drugs. 2015;13(6):3653–3671. doi: 10.3390/md13063653.
    1. Ryan J. T., Ross R. P., Bolton D., Fitzgerald G. F., Stanton C. Bioactive peptides from muscle sources: meat and fish. Nutrients. 2011;3(9):765–791. doi: 10.3390/nu3090765.
    1. Muralidharan N., Jeya Shakila R., Sukumar D., Jeyasekaran G. Skin, bone and muscle collagen extraction from the trash fish, leather jacket (Odonus niger) and their characterization. Journal of Food Science and Technology. 2013;50(6):1106–1113. doi: 10.1007/s13197-011-0440-y.
    1. Yamamoto K., Yoshizawa Y., Yanagiguchi K., Ikeda T., Yamada S., Hayashi Y. The characterization of fish (tilapia) collagen sponge as a biomaterial. International Journal of Polymer Science. 2015;2015:5. doi: 10.1155/2015/957385.957385
    1. Karim A. A., Bhat R. Fish gelatin: properties, challenges, and prospects as an alternative to mammalian gelatins. Food Hydrocolloid. 2009;23(3):563–576. doi: 10.1016/j.foodhyd.2008.07.002.
    1. Choi D. J., Choi S. M., Kang H. Y., et al. Bioactive fish collagen/polycaprolactone composite nanofibrous scaffolds fabricated by electrospinning for 3D cell culture. Journal of Biotechnology. 2015;205:47–58. doi: 10.1016/j.jbiotec.2015.01.017.
    1. Liu J., Zhang B., Song S., et al. Bovine collagen peptides compounds promote the proliferation and differentiation of MC3T3-E1 pre-osteoblasts. PloS One. 2014;9(6, article e99920) doi: 10.1371/journal.pone.0099920.
    1. Coondoo A. Cytokines in dermatology - a basic overview. Indian Journal of Dermatology. 2011;56(4):368–374. doi: 10.4103/0019-5154.84717.
    1. O'Shea J. J., Ma A., Lipsky P. Cytokines and autoimmunity. Nature Reviews. Immunology. 2002;2(1):37–45. doi: 10.1038/nri702.
    1. Sun Z., Mohamed M. A., Park S. Y., Yi T. H. Fucosterol protects cobalt chloride induced inflammation by the inhibition of hypoxia-inducible factor through PI3K/Akt pathway. International Immunopharmacology. 2015;2(2):642–647. doi: 10.1016/j.intimp.2015.09.016.
    1. Koul H. K., Pal M., Koul S. Role of p38 MAP kinase signal transduction in solid tumors. Genes & Cancer. 2013;4(9-10):342–359. doi: 10.1177/1947601913507951.
    1. Mavropoulos A., Orfanidou T., Liaskos C., et al. p38 MAPK signaling in pemphigus: implications for skin autoimmunity. Autoimmune Diseases. 2013;2013:11. doi: 10.1155/2013/728529.728529
    1. Coulthard L. R., White D. E., Jones D. L., McDermott M. F., Burchill S. A. p38MAPK: stress responses from molecular mechanisms to therapeutics. Trends in Molecular Medicine. 2009;15(8):369–379. doi: 10.1016/j.molmed.2009.06.005.
    1. Jinlian L., Yingbin Z., Chunbo W. p38 MAPK in regulating cellular responses to ultraviolet radiation. Journal of Biomedical Science. 2007;4(3):303–312. doi: 10.1007/s11373-007-9148-4.
    1. Soegaard-Madsen L., Johansen C., Iversen L., Kragballe K. Adalimumab therapy rapidly inhibits p38 mitogen-activated protein kinase activity in lesional psoriatic skin preceding clinical improvement. The British Journal of Dermatology. 2010;162(6):1216–1223. doi: 10.1111/j.1365-2133.2010.09706.x.
    1. Saklatvala J. The p38 MAP kinase pathway as a therapeutic target in inflammatory disease. Current Opinion in Pharmacology. 2004;4(4):372–377. doi: 10.1016/j.coph.2004.03.009.
    1. Saldeen J., Welsh N. p38 MAPK inhibits JNK2 and mediates cytokine-activated iNOS induction and apoptosis independently of NF-KB translocation in insulin-producing cells. European Cytokine Network. 2004;1(1):47–52.
    1. Lan A., Xu W., Zhang H., et al. Inhibition of ROS activated p38 MAPK pathway is involved in the protective effect of H2S against chemical hypoxia-induced inflammation in PC12 cells. Neurochemical Research. 2013;7(7):1454–1466. doi: 10.1007/s11064-013-1044-x.
    1. Van Laethem A., Van Kelst S., Lippens S., et al. Activation of p38 MAPK is required for Bax translocation to mitochondria, cytochrome c release and apoptosis induced by UVB irradiation in human keratinocytes. The FASEB journal. 2004;18(15):1946–1948. doi: 10.1096/fj.04-2285fje.
    1. Liu Z. G. Molecular mechanism of TNF signaling and beyond. Cell Research. 2005;15(1):24–27. doi: 10.1038/sj.cr.7290259.
    1. Aggarwal B. B., Shishodia S., Takada Y., et al. TNF blockade: an inflammatory issue. Ernst Schering Research Foundation Workshop. 2006;56(56):161–186. doi: 10.1007/3-540-37673-9_10.
    1. Sabio G., Davis R. J. TNF and MAP kinase signalling pathways. Seminars in Immunology. 2014;26(3):237–245. doi: 10.1016/j.smim.2014.02.009.
    1. Lawrence T. The nuclear factor NF-kappaB pathway in inflammation. Cold Spring Harbor Perspectives in Biology. 2009;1(6, article a001651) doi: 10.1101/cshperspect.a001651.
    1. Chai O. H., Song C. H. Role of mast cell in the late phase of contact hypersensitivity induced by trimellitic anhydride. Anatomy & Cell Biology. 2015;48(4):225–234. doi: 10.5115/acb.2015.48.4.225.
    1. Guo L., Harnedy P. A., O'Keeffe M. B., et al. Fractionation and identification of Alaska pollock skin collagen-derived mineral chelating peptides. Food Chemistry. 2015;173:536–542. doi: 10.1016/j.foodchem.2014.10.055.
    1. Huang C. Y., Wu C. H., Yang J. J., Li Y. H., Kuo J. M. Evaluation of iron-binding activity of collagen peptides prepared from the scales of four cultivated fishes in Taiwan. Journal of Food and Drug Analysis. 2015;23(4):671–678. doi: 10.1016/j.jfda.2014.06.009.

Source: PubMed

3
Abonnieren