Browse > Article
http://dx.doi.org/10.4162/nrp.2017.11.4.275

Astaxanthin induces migration in human skin keratinocytes via Rac1 activation and RhoA inhibition  

Ritto, Dakanda (Department of Pharmacology, Faculty of Science, Prince of Songkla University)
Tanasawet, Supita (Department of Anatomy, Faculty of Science, Prince of Songkla University)
Singkhorn, Sawana (Department of Pharmacology, Faculty of Science, Prince of Songkla University)
Klaypradit, Wanwimol (Department of Fishery Product, Faculty of Fishery, Kasetsart University)
Hutamekalin, Pilaiwanwadee (Department of Physiology, Faculty of Science, Prince of Songkla University)
Tipmanee, Varomyalin (Department of Biomedical Sciences, Faculty of Medicine, Prince of Songkla University)
Sukketsiri, Wanida (Department of Pharmacology, Faculty of Science, Prince of Songkla University)
Publication Information
Nutrition Research and Practice / v.11, no.4, 2017 , pp. 275-280 More about this Journal
Abstract
BACKGROUND/OBJECTIVES: Re-epithelialization has an important role in skin wound healing. Astaxanthin (ASX), a carotenoid found in crustaceans including shrimp, crab, and salmon, has been widely used for skin protection. Therefore, we investigated the effects of ASX on proliferation and migration of human skin keratinocyte cells and explored the mechanism associated with that migration. MATERIAL/METHOD: HaCaT keratinocyte cells were exposed to $0.25-1{\mu}g/mL$ of ASX. Proliferation of keratinocytes was analyzed by using MTT assays and flow cytometry. Keratinocyte migration was determined by using a scratch wound-healing assay. A mechanism for regulation of migration was explored via immunocytochemistry and western blot analysis. RESULTS: Our results suggest that ASX produces no significant toxicity in human keratinocyte cells. Cell-cycle analysis on ASX-treated keratinocytes demonstrated a significant increase in keratinocyte cell proliferation at the S phase. In addition, ASX increased keratinocyte motility across the wound space in a time-dependent manner. The mechanism by which ASX increased keratinocyte migration was associated with induction of filopodia and formation of lamellipodia, as well as with increased Cdc42 and Rac1 activation and decreased RhoA activation. CONCLUSIONS: ASX stimulates the migration of keratinocytes through Cdc42, Rac1 activation and RhoA inhibition. ASX has a positive role in the re-epithelialization of wounds. Our results may encourage further in vivo and clinical study into the development of ASX as a potential agent for wound repair.
Keywords
Carotenoids; cell movement; wound healing; re-epithelialization;
Citations & Related Records
Times Cited By KSCI : 1  (Citation Analysis)
연도 인용수 순위
1 Xu Y, Zhang J, Jiang W, Zhang S. Astaxanthin induces angiogenesis through Wnt/$\beta$-catenin signaling pathway. Phytomedicine 2015;22: 744-51.   DOI
2 Schlessinger K, Hall A, Tolwinski N. Wnt signaling pathways meet Rho GTPases. Genes Dev 2009;23:265-77.   DOI
3 Sangsuriyawong A, Limpawattana M, Siriwan D, Kaewnern P, Klaypradit W. Effects of phospholipid concentration and mixing methods on properties of astaxanthin extract-loaded liposomes. J Agric 2016;32:421-33.
4 Taksima T, Limpawattana M, Klaypradit W. Astaxanthin encapsulated in beads using ultrasonic atomizer and application in yogurt as evaluated by consumer sensory profile. LWT Food Sci Technol 2015;62:431-7.   DOI
5 Kuedo Z, Sangsuriyawong A, Klaypradit W, Tipmanee V, Chonpathompikunlert P. Effects of astaxanthin from Litopenaeus vannamei on carrageenan-induced edema and pain behavior in mice. Molecules 2016;21:382.   DOI
6 Santoro MM, Gaudino G. Cellular and molecular facets of keratinocyte reepithelization during wound healing. Exp Cell Res 2005;304: 274-86.   DOI
7 Harper D, Young A, McNaught C. The physiology of wound healing. Surgery 2014;32:445-50.
8 Kim JH, Nam SW, Kim BW, Choi W, Lee JH, Kim WJ, Choi YH. Astaxanthin improves stem cell potency via an increase in the proliferation of neural progenitor cells. Int J Mol Sci 2010;11: 5109-19.   DOI
9 Hodge RG, Ridley AJ. Regulating Rho GTPases and their regulators. Nat Rev Mol Cell Biol 2016;17:496-510.   DOI
10 Fukata M, Nakagawa M, Kaibuchi K. Roles of Rho-family GTPases in cell polarisation and directional migration. Curr Opin Cell Biol 2003;15:590-7.   DOI
11 Raftopoulou M, Hall A. Cell migration: Rho GTPases lead the way. Dev Biol 2004;265:23-32.   DOI
12 Li J, Zhang S, Soto X, Woolner S, Amaya E. ERK and phosphoinositide 3-kinase temporally coordinate different modes of actin-based motility during embryonic wound healing. J Cell Sci 2013;126:5005-17.   DOI
13 Zhang Z, Yang M, Chen R, Su W, Li P, Chen S, Chen Z, Chen A, Li S, Hu C. IBP regulates epithelial-to-mesenchymal transition and the motility of breast cancer cells via Rac1, RhoA and Cdc42 signaling pathways. Oncogene 2014;33:3374-82.   DOI
14 Rorth P. Collective cell migration. Annu Rev Cell Dev Biol 2009;25: 407-29.   DOI
15 Zhao X, Guan JL. Focal adhesion kinase and its signaling pathways in cell migration and angiogenesis. Adv Drug Deliv Rev 2011;63:610-5.   DOI
16 Gammone MA, Riccioni G, D'Orazio N. Marine carotenoids against oxidative stress: effects on human health. Mar Drugs 2015;13:6226-46.   DOI
17 Gurtner GC, Werner S, Barrandon Y, Longaker MT. Wound repair and regeneration. Nature 2008;453:314-21.   DOI
18 Salhia B, Rutten F, Nakada M, Beaudry C, Berens M, Kwan A, Rutka JT. Inhibition of Rho-kinase affects astrocytoma morphology, motility, and invasion through activation of Rac1. Cancer Res 2005;65:8792-800.   DOI
19 Xu J, Wang F, Van Keymeulen A, Herzmark P, Straight A, Kelly K, Takuwa Y, Sugimoto N, Mitchison T, Bourne HR. Divergent signals and cytoskeletal assemblies regulate self-organizing polarity in neutrophils. Cell 2003;114:201-14.   DOI
20 Petpiroon N, Suktap C, Pongsamart S, Chanvorachote P, Sukrong S. Kaempferol-3-O-rutinoside from Afgekia mahidoliae promotes keratinocyte migration through FAK and Rac1 activation. J Nat Med 2015;69:340-8.   DOI
21 Wadman M. Scar prevention: the healing touch. Nature 2005;436: 1079-80.   DOI
22 Haase I, Evans R, Pofahl R, Watt FM. Regulation of keratinocyte shape, migration and wound epithelialization by IGF-1- and EGF-dependent signalling pathways. J Cell Sci 2003;116:3227-38.   DOI
23 Enciso JM, Konecny CM, Karpen HE, Hirschi KK. Endothelial cell migration during murine yolk sac vascular remodeling occurs by means of a Rac1 and FAK activation pathway in vivo. Dev Dyn 2010;239:2570-83.   DOI
24 Hsu CL, Muerdter CP, Knickerbocker AD, Walsh RM, Zepeda-Rivera MA, Depner KH, Sangesland M, Cisneros TB, Kim JY, Sanchez-Vazquez P, Cherezova L, Regan RD, Bahrami NM, Gray EA, Chan AY, Chen T, Rao MY, Hille MB. Cdc42 GTPase and Rac1 GTPase act downstream of p120 catenin and require GTP exchange during gastrulation of zebrafish mesoderm. Dev Dyn 2012;241:1545-61.   DOI
25 Burridge K, Chrzanowska-Wodnicka M. Focal adhesions, contractility, and signaling. Annu Rev Cell Dev Biol 1996;12:463-518.   DOI
26 Sepp KJ, Auld VJ. RhoA and Rac1 GTPases mediate the dynamic rearrangement of actin in peripheral glia. Development 2003; 130:1825-35.   DOI
27 Nobes CD, Hall A. Rho GTPases control polarity, protrusion, and adhesion during cell movement. J Cell Biol 1999;144:1235-44.   DOI
28 Rottner K, Hall A, Small JV. Interplay between Rac and Rho in the control of substrate contact dynamics. Curr Biol 1999;9:640-8.   DOI
29 Kidd P. Astaxanthin, cell membrane nutrient with diverse clinical benefits and anti-aging potential. Altern Med Rev 2011;16:355-64.
30 Naguib YM. Antioxidant activities of astaxanthin and related carotenoids. J Agric Food Chem 2000;48:1150-4.   DOI
31 Ranga Rao A, Baskaran V, Sarada R, Ravishankar GA. In vivo bioavailability and antioxidant activity of carotenoids from microalgal biomass: a repeated dose study. Food Res Int 2013; 54:711-7.   DOI
32 Praveenkumar R, Gwak R, Kang M, Shim TS, Cho S, Lee J, Oh YK, Lee K, Kim B. Regenerative astaxanthin extraction from a single microalgal (Haematococcus pluvialis) cell using a gold nano-scalpel. ACS Appl Mater Interfaces 2015;7:22702-8.   DOI
33 Choi SK, Park YS, Choi DK, Chang HI. Effects of astaxanthin on the production of NO and the expression of COX-2 and iNOS in LPS-stimulated BV2 microglial cells. J Microbiol Biotechnol 2008; 18:1990-6.
34 Kishimoto Y, Tani M, Uto-Kondo H, Iizuka M, Saita E, Sone H, Kurata H, Kondo K. Astaxanthin suppresses scavenger receptor expression and matrix metalloproteinase activity in macrophages. Eur J Nutr 2010;49:119-26.   DOI
35 Camera E, Mastrofrancesco A, Fabbri C, Daubrawa F, Picardo M, Sies H, Stahl W. Astaxanthin, canthaxanthin and $\beta$-carotene differently affect UVA-induced oxidative damage and expression of oxidative stress-responsive enzymes. Exp Dermatol 2009;18:222-31.   DOI
36 Hama S, Takahashi K, Inai Y, Shiota K, Sakamoto R, Yamada A, Tsuchiya H, Kanamura K, Yamashita E, Kogure K. Protective effects of topical application of a poorly soluble antioxidant astaxanthin liposomal formulation on ultraviolet-induced skin damage. J Pharm Sci 2012;101:2909-16.   DOI
37 Mizuta M, Hirano S, Hiwatashi N, Tateya I, Kanemaru S, Nakamura T, Ito J. Effect of astaxanthin on vocal fold wound healing. Laryngoscope 2014;124:E1-7.   DOI