MATRIX METALLOPROTEINASES AS PATHOGENETIC AND DIAGNOSTIC BIOMARKERS FOR SKIN SCARS

Download full text PDF
Issue: 
3
Year: 
2017

E. Luchina, Candidate of Medical Sciences S.M. Kirov Military Medical Academy, Saint Petersburg

To develop optimal treatments for skin scarring changes, it is necessary to understand the pathogenesis of the wound process that is a complex set of biological reactions.

Keywords: 
dermatology
scarring tissue
biomarkers
matrix metalloproteinases



It appears your Web browser is not configured to display PDF files. Download adobe Acrobat или click here to download the PDF file.

References: 
  1. Sten'ko A.G., Kruglova L.S., Shmatova A.A. i dr. Konservativnoe lechenie formirujuschihsja rubtsov: obzor sovremennyh tehnologij // Vestnik esteticheskoj meditsiny. – 2014; 13 (2): 42–50.
  2. Tarasenkova M.S., Jutskovskaja Ja.A., Naumchik G.A. i dr. Sovremennye tsmetody profilaktiki postoperatsionnyh patologicheskih rubtsov kozhi // Eksperim. i klin. dermatokosmetol. – 2010; 3: 50–4.
  3. Roh Y., Seo C., Jang K. Effects of a skin rehabilitation nursing program on skin status, depression, and burn-specific health in burn survivors // Rehabil. Nurs. – 2010; 35 (2): 65–9.
  4. Mamalis A., Jagdeo J. Light-emitting diode-generated red light inhibits keloid fibroblast proliferation // Dermatol. Surg. – 2015; 41 (1): 35–9.
  5. Wang A., Zhang W., Liang F. et al. Pre-expanded thoracodorsal artery perforator-based flaps for repair of severe scarring in cervicofacial regions // J. Reconstr. Microsurg. – 2014; 30 (8): 539–46.
  6. Gill S., Parks W. Metalloproteinases and their inhibitors: regulators of wound healing // Int. J. Biochem. Cell Biol. – 2008; 40: 1334–47.
  7. Gershtejn E.S., Levkina N.V., Digaeva M.A. i dr. Matriksnye metalloproteinazy-2, -7, -9 i tkanevoj ingibitor metalloproteinaz 1-go tipa v opuholjah i syvorotke krovi bol'nyh s novoobrazovanijami jaichnikov // Bjul. eksperim. biol. i meditsiny. – 2010; 149 (5): 562–5.
  8. Nissi R., Talvensaari-Mattila A., Kotila V. et al. Circulating matrix metalloproteinase MMP-9 and MMP-2/TIMP-2 complex are associated with spontaneous early pregnancy failure // Reprod. Biol. Endocrinol. – 2013; 15 (11): 2
  9. Cho S., Ryu D., Lee S. et al Scar characteristics and treatment expectations: a survey of 589 patients // Cosmet. Laser Ther. – 2009; 11 (4): 224–8.
  10. Wagner W., Alfrink M., Micke O. Results of prophylactic irradiation in patients with resected keloids a retrospective analysis // Acta. Oncol. – 2000; 39 (2): 217–20.
  11. Margolina A.A., Ernandes E.I., Zajkina O.E. Novaja kosmetologija / M.: Kosmetika i meditsina, 2002; 208 s.
  12. Matytsin V.O., Miheeva N.V. Metody instrumental'noj diagnostiki i funktsional'nogo sostojanija kozhi // Natural'naja farmakol. i kosmetol. – 2005; 2: 35–7.
  13. Snarskaja E.S. Kompleksnaja terapija esteticheskih defektov kozhi v rezul'tate patologicheskogo fibrogeneza // Dermatologija. Prilozhenie k zhurnalu Consilium Medicum. – 2013; 2–3: 15–20.
  14. Artykov K.P., Saidov M.S., Muhamadieva K.M. Vlijanie immunomodulirujuschej terapii na rezul'taty hirurgicheskoj korrektsii kelloidnyh rubtsov kozhi // Doklady Akademii nauk Respubliki Tadzhikistan. – 2014; 57 (2): 164–9.
  15. Chen J., Wang J., Zhuang H. Influence of substance P on the proliferation and apoptosis of fibroblasts of pathological scars // Zhonghua Shao Shang ZaZhi. – 2006; 22 (4): 277–80.
  16. Sauder D.N. Cutaneous immunobiology // Ann. Dermatol. Venereol. – 2002; 129: 274–83.
  17. Wolfram D., Tzankov A., Polzl P. et al. Hypertrophic scars and keloids. A review of their pathophysiology, risk factors, and therapeutic management // Dermatol. Surg. – 2009; 35: 171–81.
  18. Eto H., Suga H., Aoi N. et al. Therapeutic potential of fibroblast growth factor-2 for hypertrophic scars: upregulation of MMP-1 and HGF expression // Lab. Invest. – 2012; 92: 214–23.
  19. Katunina A.I., Gershtejn E.S., Tereshkina I.V. i dr. Matriksnye metalloproteinazy v opuholjah bol'nyh rakom molochnoj zhelezy // Klin. lab. diagnostika. – 2010; 9: 27–7a.
  20. Ortak H., Demir S., Ateş Ö. et al. The role of MMP2 (–1306C>T) and TIMP2(–418G>C) promoter variants in age-related macular degeneration // Ophthalmic Genet. – 2013; 34 (4): 217–22.
  21. Imaizumi R., Akasaka Y., Inomata N. et al. Promoted activation of matrix metalloproteinase (MMP)-2 in keloid fibroblasts and increased expression of MMP-2 in collagen bundle regions: implications for mechanisms of keloid progression // Histopathology. – 2009; 54: 722–30.
  22. Tandara A., Mustoe T. MMP- and TIMP-secretion by human cutaneous keratinocytes and fibroblasts – impact of coculture and hydration // J. Plast. Reconstr. Aesthet. Surg. – 2011; 64: 108–16.
  23. Ulrich D., Ulrich F., Unglaub F. et al. Matrix metalloproteinases and tissue inhibitors of metalloproteinases in patients with different types of scars and keloids // J. Plast. Reconstr. Aesthet. Surg. – 2010; 63: 1015–21.
  24. Simon F., Bergeron D., Larochelle S. et al. Enhanced secretion of TIMP-1 by human hypertrophic scar keratinocytes could contribute to fibrosis // Burns. – 2012; 38: 421–7.
  25. Taghiabadi E., Mohammadi P., Aghdami N. et al. Treatment of Hypertrophic Scar in Human with Autologous Transplantation of Cultured Keratinocytes and Fibroblasts along with Fibrin Glue // Cell J. – 2015; 17 (1): 49–58.
  26. Sadick H., Herberger A., Riedel K. et al. TGF-beta1 antisense therapy modulates expression of matrix metalloproteinases in keloid-derived fibroblasts // Int. J. Mol. Med. – 2008; 22: 55–60.
  27. Fujiwara M., Muragaki Y., Ooshima A. Keloid-derived fibroblasts show increased secretion of factors involved in collagen turnover and depend on matrix metalloproteinase for migration // Br. J. Dermatol. – 2005; 153: 295–300.
  28. Uchida G., Yoshimura K., Kitano Y. et al. Tretinoin reverses upregulation of matrix metalloproteinase-13 in human keloid derived fibroblasts // Exp. Dermatol. – 2003; 12 (Suppl. 2): 35–42.
  29. Chavez-Munoz C., Hartwell R., Jalili R. et al. Application of an indoleamine 2,3-dioxygenase-expressing skin substitute improves scar formation in a fibrotic animal model // J. Invest. Dermatol. – 2012; 132: 1501–5.
  30. Li Y., Kilani R., Rahmani-Neishaboor E. et al. Kynurenine increases matrix metalloproteinase-1 and -3 expression in cultured dermal fibroblasts and improves scarring in vivo // J. Invest. Dermatol. – 2014; 134: 643–50.
  31. Wei Y., Yan X., Ma L. et al. Oleanolic acid inhibits hypertrophic scarring in the rabbit ear model // Clin. Exp. Dermatol. – 2011; 36: 528–33.
  32. Kuo Y., Wu W., Jeng S. et al. Suppressed TGF-beta1 expression is correlated with up-regulation of matrix metalloproteinase-13 in keloid regression after flashlamp pulsed-dye laser treatment // Lasers Surg. Med. – 2005; 36: 38–42.
  33. Rahmani-Neishaboor E., Yau F., Jalili R. et al. Improvement of hypertrophic scarring by using topical anti-fibrogenic/anti-inflammatory factors in a rabbit ear model // Wound Repair Regen. – 2010; 18: 401–8.
  34. Djafarzadeh R., Conrad C., Notohamiprodjo S. et al. Cell surface engineering using glycosylphosphatidylinositol anchored tissue inhibitor of matrix metalloproteinase-1 stimulates cutaneous wound healing // Wound Repair Regen. – 2014; 22: 70–6.
  35. Stuart K., Paderi J., Snyder P. et al. Collagen-binding peptidoglycans inhibit MMP mediated collagen degradation and reduce dermal scarring // PLoS One. – 2011; 6: e22139.
  36. Ghahary A., Karimi-Busheri F., Marcoux Y. et al. Keratinocyte-releasable stratifin functions as a potent collagenase-stimulating factor in fibroblasts // J. Invest. Dermatol. – 2004; 122: 1188–97.
  37. Rahmani-Neishaboor E., Jackson J., Burt H. et al. Composite hydrogel formulations of stratifin to control MMP-1 expression in dermal fibroblasts // Pharm. Res. – 2009; 26: 002–2014.
  38. Lee W., Ahn H., Roh H. et al. Decorin-expressing adenovirus decreases collagen synthesis and upregulates MMP expression in keloid fibroblasts and keloid spheroids // Exp. Dermatol. – 2015; 24 (8): 591–7.