Skip to main content
Log in

Effect of Palmitoyl-Pentapeptide (Pal-KTTKS) on Wound Contractile Process in Relation with Connective Tissue Growth Factor and α-Smooth Muscle Actin Expression

  • Original Article
  • Published:
Tissue Engineering and Regenerative Medicine Aims and scope

Abstract

To evaluate whether Palmitoyl-pentapeptide (Pal-KTTKS), a lipidated subfragment of type 1 pro-collagen (residues 212–216), plays a role in fibroblast contractility, the effect of Pal-KTTKS on the expression of pro-fibrotic mediators in hypertropic scarring were investigated in relation with trans-differentiation of fibroblast to myofibroblast, an icon of scar formation. α-SMA was visualized by immunofluorescence confocal microscopy with a Cy-3-conjugated monoclonal antibody. The extent of α-SMA-positive fibroblasts was determined in collagen lattices and in cell culture study. Pal-KTTKS (0–0.5 µM) induced CTGF and α-SMA protein levels were determined by western blot analysis and fibroblast contractility was assessed in three-dimensional collagen lattice contraction assay. In confocal analysis, fibroblasts were observed as elongated and spindle shapes while myofibroblast observed as squamous, enlarged cells with pronounced stress fibers. Without Pal-KTTKS treatment, three quarters of the fibroblasts differentiates into the myofibroblast; α-SMA-positive stress fibers per field decreased twofold with 0.1 µM Pal-KTTKS treatment (75 ± 7.1 vs 38.6 ± 16.1%, n = 3, p < 0.05). The inhibitory effect was not significant in 0.5 µM Pal-KTTKS treatment. Stress fiber level and collagen contractility correlates with α-SMA expression level. In conclusion, Pal-KTTKS (0.1 µM) reduces α-SMA expression and trans-differentiation of fibroblasts to myofibroblast. The degree of reduction is dose-dependent. An abundance of myofibroblast and fibrotic scarring is correlated with excessive levels of α-SMA and collagen contractility. Delicate balance between the wound healing properties and pro-fibrotic abilities of pentapeptide KTTKS should be considered for selecting therapeutic dose for scar prevention.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. Katayama K, Armendariz Borunda J, Raghow R. A pentapeptide from type 1 procollagen promotes extracellular matrix production. J Biol Chem. 1993;268:9941–4.

    PubMed  CAS  Google Scholar 

  2. Castelletto V, Hamley IW, Perez J, Abezgauz L, Danino D. Fibrillar superstructure from extended nanotapes formed by a collagen-stimulating peptide. Chem Commun. 2010;46:9185–87.

    Article  CAS  Google Scholar 

  3. Tsai WC, Hsu CC, Chung CY, Lin MS, Li MS, Pang JH. The pentapeptide KTTKS promoting the expressions of type 1 collagen and transforming growth factor-β of tendon cells. J Orthop Res. 2007;25:1629–34.

    Article  PubMed  CAS  Google Scholar 

  4. Jones RR, Castelletto V, Connon CJ, Hamley IW. Collagen stimulating effect of peptide amphiphile C-16-KTTKS on human fibroblasts. Mol Pharm. 2013;10:1063–9.

    Article  PubMed  CAS  Google Scholar 

  5. Juan FM, Beatriu E, Maria DS-M, Marta TS, Ian WH, Ashkan D, et al. Self-assembly of a peptide amphiphile: transition from nanotape fibrils to micelles. Soft Matter. 2013;9:3558–64.

    Article  CAS  Google Scholar 

  6. Farwick M, Grether-Beck S, Marini A, Maczkiewitz U, Lange J, Kohler T, et al. Bioactive tetrapeptide GEKG boosts extracellular matrix formation: in vitro and in vivo molecular and clinical proof. Exp Dermatol. 2011;20:600–13.

    Article  Google Scholar 

  7. Maquart FX, Pasco S, Ramont L, Hornebeck W, Monboisse JC. An introduction to matrikines: extracellular matrix-derived peptides which regulate cell activity implication in tumor invasion. Crit Rev Oncol Hematol. 2004;49:199–202.

    Article  PubMed  Google Scholar 

  8. Roanne RJ, Valeria C, Che JC, Ian WH. Collagen stimulating effect of peptide amphiphile C 16-KTTKS on human fibroblasts. Mol Pharm. 2013;10:1063–9.

    Article  CAS  Google Scholar 

  9. Abu Samah NH, Heard CM. Topically applied KTTKS: a review. Int J Cosmet Sci. 2011;33:483–90.

    Article  PubMed  CAS  Google Scholar 

  10. Reid R, Mogford JE, Butt R, Miller AG, Mustoe T. Inhibition of procollagen C-proteinase reduces scar hyperthrophy in a rabbit model of cutaneous scarring. Wound Rep Regen. 2006;14:138–41.

    Article  Google Scholar 

  11. Yang M, Huang H, Li J, Huang W, Wang H. Connective tissue growth factor increases matrix metalloproteinase-2 and suppresses tissue inhibitor of matrix metalloproteinase-2 production by cultured renal interstitial fibroblasts. Wound Rep Regen. 2007;15:817–24.

    Article  Google Scholar 

  12. Alfaro MP, Deskins DL, Wallus M, DasGupta J, Davidson JM, Nanney LB, et al. A physiological role for connective tissue growth factor in early wound healing. Lab Invest. 2013;93:81–95.

    Article  PubMed  CAS  Google Scholar 

  13. Frazier K, Williams S, Kothapalli D, Klapper H, Grotendors GR. Stimulation of fibroblast cell growth, matrix production and granulation tissue formation by CTGF. J Invest Dermatol. 1996;107:404–11.

    Article  PubMed  CAS  Google Scholar 

  14. Leask A, Holmes A, Abraham DJ. Connective tissue growth factor: a new and important player in the pathogenesis of fibrosis. Curr Rheumatol Rep. 2002;4:136–42.

    Article  PubMed  Google Scholar 

  15. Sonnylal S, Shi-wen X, Leoni P, Naff K, Van Pelt C, Nakamura H, et al. Selective expression of CTGF in fibroblasts in vivo promotes systemic tissue fibrosis. Arthritis Rheum. 2010;62:1523–32.

    Article  PubMed  CAS  Google Scholar 

  16. Hinz B. Formation and function of the myofibroblast during tissue repair. J Invest Dermatol. 2007;127:526–37.

    Article  PubMed  CAS  Google Scholar 

  17. Hinz B, Celetta G, Tomasek JJ, Gabbiani G, Chaponnier C. Alpha-smooth muscle actin expression upregulates fibroblast contractile activity. Mol Biol Cell. 2001;12:2730–41.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Gabbiani G. The myofibroblast in wound healing and fibrocontractive diseases. J Pathol. 2003;200:500–3.

    Article  PubMed  CAS  Google Scholar 

  19. Daniels JT, Schultz GS, Blalock TD, Garrett Q, Grotendorst GR, Dean NM, et al. Mediation of transforming growth factor-β1-stimulated matrix contraction by fibroblasts: a role for CTGF in contractile scarring. Am J Pathol. 2003;163:2043–52.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Desai VD, Hsia HC, Schwarzbauer JE. Reversible modulation of myofibroblast differentiation in adipose-derived mesenchymal stem cells. PLoS ONE. 2014;9:e86865.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Evans RA, Tian YC, Steadman R, Phillips AO. TGF-1-mediated fibroblast-myofibroblast terminal differentiation—the role of Smad proteins. Exp Cell Res. 2003;282:90–100.

    Article  PubMed  CAS  Google Scholar 

  22. Moon HK, Yong HY, Cho Lee AR. Optimum scratch assay condition to evaluate CTGF expression for anti-scar therapy. Arch Pharm Res. 2012;35:383–8.

    Article  PubMed  CAS  Google Scholar 

  23. Tang J, Sato T. Effect of type I collagen derivative from Tilapia Scale on odontobalst-like cells. Tissue Eng Regen Med. 2015;12:231–8.

    Article  CAS  Google Scholar 

  24. Diegelman RF, Evans MC. Wound Healing: an overview of acute, Fibrotic and delayed healing. Front Biosci. 2004;9:283–9.

    Article  Google Scholar 

  25. Boris H, Giuseppe C, James JT, Giulio G, Christine C. Alpha-smooth muscle actin expression upregulates fibroblast contractile activity. Mol Biol Cell. 2001;12:2730–41.

    Article  Google Scholar 

  26. Rinkevich Y, Walmsley G, Hu M, Maan A, Newman A, Drukker M, et al. Identification and isolation of a dermal lineage with intrinsic fibrogenic potential. Science 2015;348(6232). doi:10.1126/science.aaa2151.

  27. Mas-Chamberlin C, Mondon P, Peschard O, Lintner K. Matrikine technology and barrier repair: the ultimate in anti-age skin care? Cosmetic Sci Technol 2004;53.

  28. Dehsorkhi A, Castelletto V, Hamley IW. Self-assembling amphiphilic peptides. J Pept Sci. 2014;20:453–67.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Maquart FX, Bellon G, Pasco S, Monboisse JC. Matrikines in the regulation of extracellular matrix degradation. Biochimie. 2005;87:353–60.

    Article  PubMed  CAS  Google Scholar 

  30. Choi YL, Park EJ, Kim E, Na DH, Shin YH. Dermal stability and in vitro skin permeation of collagen pentapeptides (KTTKS). Biomol Ther. 2014;22:321–7.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the Duksung Women’s University Research Grants 2014. We thank Prof. Kyung Chan Park at Department of Dermatology, School of Medicine, Seoul National University for his valuable discussion and for providing primary human dermal fibroblasts.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ae-Ri Cho Lee.

Ethics declarations

Conflict of interest statement

The authors declare no conflict of interest with any person or any organization.

Ethical statement

There are no animal experiments carried out for this study.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Park, H., An, E. & Cho Lee, AR. Effect of Palmitoyl-Pentapeptide (Pal-KTTKS) on Wound Contractile Process in Relation with Connective Tissue Growth Factor and α-Smooth Muscle Actin Expression. Tissue Eng Regen Med 14, 73–80 (2017). https://doi.org/10.1007/s13770-016-0017-y

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13770-016-0017-y

Keywords

Navigation