Angiogenic properties of dehydrated human amnion/chorion allografts: therapeutic potential for soft tissue repair and regeneration

Thomas J Koob, Jeremy J Lim, Michelle Massee, Nicole Zabek, Robert Rennert, Geoffrey Gurtner, William W Li, Thomas J Koob, Jeremy J Lim, Michelle Massee, Nicole Zabek, Robert Rennert, Geoffrey Gurtner, William W Li

Abstract

Background: Chronic wounds are associated with a number of deficiencies in critical wound healing processes, including growth factor signaling and neovascularization. Human-derived placental tissues are rich in regenerative cytokines and have been shown in randomized clinical trials to be effective for healing chronic wounds. In this study, PURION® Processed (MiMedx Group, Marietta, GA) dehydrated human amnion/chorion membrane tissue allografts (dHACM, EpiFix®, MiMedx) were evaluated for properties to support wound angiogenesis.

Methods: Angiogenic growth factors were identified in dHACM tissues using enzyme-linked immunosorbent assays (ELISAs), and the effects of dHACM extract on human microvascular endothelial cell (HMVEC) proliferation and production of angiogenic growth factors was determined in vitro. Chemotactic migration of human umbilical vein endothelial cells (HUVECs) toward pieces of dHACM tissue was determined using a standard in vitro transwell assay. Neovascularization of dHACM in vivo was determined utilizing a murine subcutaneous implant model.

Results: Quantifiable levels of the angiogenic cytokines angiogenin, angiopoietin-2 (ANG-2), epidermal growth factor (EGF), basic fibroblast growth factor (bFGF), heparin binding epidermal growth factor (HB-EGF), hepatocyte growth factor (HGF), platelet derived growth factor BB (PDGF-BB), placental growth factor (PlGF), and vascular endothelial growth factor (VEGF) were measured in dHACM. Soluble cues promoted HMVEC proliferation in vitro and increased endogenous production of over 30 angiogenic factors by HMVECs, including granulocyte macrophage colony-stimulating factor (GM-CSF), angiogenin, transforming growth factor β3 (TGF-β3), and HB-EGF. 6.0 mm disks of dHACM tissue were also found to recruit migration of HUVECs in vitro. Moreover, subcutaneous dHACM implants displayed a steady increase in microvessels over a period of 4 weeks, indicative of a dynamic intra-implant neovascular process.

Conclusions: TAKEN TOGETHER, THESE RESULTS DEMONSTRATE THAT DHACM GRAFTS: 1) contain angiogenic growth factors retaining biological activity; 2) promote amplification of angiogenic cues by inducing endothelial cell proliferation and migration and by upregulating production of endogenous angiogenic growth factors by endothelial cells; and 3) support the formation of blood vessels in vivo. dHACM grafts are a promising wound care therapy with the potential to promote revascularization and tissue healing within poorly vascularized, non-healing wounds.

Keywords: Amnion; Amnion/chorion grafts; Angiogenesis; Chorion; Chronic wounds; Endothelial cells; Growth factors; Soft tissue regeneration; VEGF; Wound healing; dHACM.

Figures

Figure 1
Figure 1
Effects of extracts of dHACM on microvascular endothelial cell proliferation in vitro. dHACM extracts promoted proliferation of HMVECs over controls; however, no dose response was observed at these concentrations. The p values shown indicate statistical significance from their respective controls. Values shown are means ± standard deviation (n = 5).
Figure 2
Figure 2
Change in growth factor production by human microvascular endothelial cells when cultured in the presence of varying concentrations of dHACM extract. Endothelial cells increased production of a number of angiogenic factors when cultured in the presence of dHACM extract, compared to untreated cells cultured without extract (green/blue). A few regulatory factors were also down regulated in the presence of extract (red/yellow), while many factors remained unchanged (black), relative to untreated controls.
Figure 3
Figure 3
Average number of migrated human umbilical vein endothelial cells (HUVECs) per field of view in response to dehydrated human amnion/chorion tissue allografts (dHACM). Representative micrographs and cell counts indicated that greater migration was observed in response to larger samples, relative to their smaller counterparts. HUVEC migration in complete medium was significantly greater than all other samples (p ≤ 0.05). * indicates significantly greater migration than basal medium and 2.0 mm groups (p ≤ 0.05). Scale bar – 200 μm.
Figure 4
Figure 4
Neovascular response following in vivo dHACM implantation. An increasing number of CD31 positive microvessels were seen within the implanted dHACM over time, ultimately reaching the level of normal and previously injured skin by day 28. (A) CD31 positive microvessels in dHACM over 28 days were stained green, while cell nuclei were counterstained blue with DAPI. (B) Vessel counts demonstrated an increase in vascularization approaching that of healthy and healing skin after 28 days. Scale bar – 50 μm.

References

    1. Bauer SM, Bauer RJ, Velazquez OC. Angiogenesis, vasculogenesis, and induction of healing in chronic wounds. Vasc Endovascular Surg. 2005;39:293–306. doi: 10.1177/153857440503900401.
    1. Tonnesen MG, Feng X, Clark RA. Angiogenesis in wound healing. J Investig Dermatol Symp Proc. 2000;5:40–46. doi: 10.1046/j.1087-0024.2000.00014.x.
    1. Blakytny R, Jude E. The molecular biology of chronic wounds and delayed healing in diabetes. Diabet Med. 2006;23:594–608. doi: 10.1111/j.1464-5491.2006.01773.x.
    1. Kim KA, Shin YJ, Kim JH, Lee H, Noh SY, Jang SH, Bae ON. Dysfunction of endothelial progenitor cells under diabetic conditions and its underlying mechanisms. Arch Pharm Res. 2012;35:223–234. doi: 10.1007/s12272-012-0203-y.
    1. Mermet I, Pottier N, Sainthillier JM, Malugani C, Cairey-Remonnay S, Maddens S, Riethmuller D, Tiberghien P, Humbert P, Aubin F. Use of amniotic membrane transplantation in the treatment of venous leg ulcers. Wound Repair Regen. 2007;15:459–464. doi: 10.1111/j.1524-475X.2007.00252.x.
    1. Subrahmanyam M. Amniotic membrane as a cover for microskin grafts. Br J Plast Surg. 1995;48:477–478. doi: 10.1016/0007-1226(95)90123-X.
    1. Gruss JS, Jirsch DW. Human amniotic membrane: a versatile wound dressing. Can Med Assoc J. 1978;118:1237–1246.
    1. John T. Human amniotic membrane transplantation: past, present, and future. Ophthalmol Clin North Am. 2003;16:43–65. doi: 10.1016/S0896-1549(02)00110-4. vi.
    1. Kubo M, Sonoda Y, Muramatsu R, Usui M. Immunogenicity of human amniotic membrane in experimental xenotransplantation. Invest Ophthalmol Vis Sci. 2001;42:1539–1546.
    1. Ueta M, Kweon MN, Sano Y, Sotozono C, Yamada J, Koizumi N, Kiyono H, Kinoshita S. Immunosuppressive properties of human amniotic membrane for mixed lymphocyte reaction. Clin Exp Immunol. 2002;129:464–470. doi: 10.1046/j.1365-2249.2002.01945.x.
    1. Dua HS, Gomes JA, King AJ, Maharajan VS. The amniotic membrane in ophthalmology. Surv Ophthalmol. 2004;49:51–77. doi: 10.1016/j.survophthal.2003.10.004.
    1. Toda A, Okabe M, Yoshida T, Nikaido T. The potential of amniotic membrane/amnion-derived cells for regeneration of various tissues. J Pharmacol Sci. 2007;105:215–228. doi: 10.1254/jphs.CR0070034.
    1. Zelen CM, Serena TE, Denoziere G, Fetterolf DE. A prospective randomised comparative parallel study of amniotic membrane wound graft in the management of diabetic foot ulcers. Int Wound J. 2013;10:502–507.
    1. Forbes J, Fetterolf DE. Dehydrated amniotic membrane allografts for the treatment of chronic wounds: a case series. J Wound Care. 2012;21:290. 292, 294–296.
    1. Sheikh ES, Sheikh ES, Fetterolf DE. Use of dehydrated human amniotic membrane allografts to promote healing in patients with refractory non healing wounds. Int Wound J. 2013. doi:10.1111/iwj.12035.
    1. Lopez-Valladares MJ, Teresa Rodriguez-Ares M, Tourino R, Gude F, Teresa Silva M, Couceiro J. Donor age and gestational age influence on growth factor levels in human amniotic membrane. Acta Ophthalmol. 2010;88:e211–216. doi: 10.1111/j.1755-3768.2010.01908.x.
    1. Russo A, Bonci P, Bonci P. The effects of different preservation processes on the total protein and growth factor content in a new biological product developed from human amniotic membrane. Cell Tissue Bank. 2012;13:353–361. doi: 10.1007/s10561-011-9261-5.
    1. Koizumi NJ, Inatomi TJ, Sotozono CJ, Fullwood NJ, Quantock AJ, Kinoshita S. Growth factor mRNA and protein in preserved human amniotic membrane. Curr Eye Res. 2000;20:173–177. doi: 10.1076/0271-3683(200003)2031-9FT173.
    1. Daniel J, Tofe R, Spencer R, Russo J. Placental tissue grafts. U.S. Patent. 2008. p. 8,357,403.
    1. Daniel J. Placental tissue grafts. U.S. Patent. 2007. p. 8,372,437.
    1. Daniel J, Tofe R, Spencer R, Russo J. Placental tissue grafts. U.S. Patent. 2012. p. 8,409,626.
    1. Koob TJ, Rennert R, Zabek N, Massee M, Lim JJ, Temenoff JS, Li WW, Gurtner G. Biological properties of dehydrated human amnion/chorion composite graft: implications for chronic wound healing. Int Wound J. 2013;10:493–500.
    1. Werner S, Grose R. Regulation of wound healing by growth factors and cytokines. Physiol Rev. 2003;83:835–870.
    1. Wieman TJ, Smiell JM, Su Y. Efficacy and safety of a topical gel formulation of recombinant human platelet-derived growth factor-BB (becaplermin) in patients with chronic neuropathic diabetic ulcers. A phase III randomized placebo-controlled double-blind study. Diabetes Care. 1998;21:822–827. doi: 10.2337/diacare.21.5.822.
    1. Smiell JM, Wieman TJ, Steed DL, Perry BH, Sampson AR, Schwab BH. Efficacy and safety of becaplermin (recombinant human platelet-derived growth factor-BB) in patients with nonhealing, lower extremity diabetic ulcers: a combined analysis of four randomized studies. Wound Repair Regen. 1999;7:335–346. doi: 10.1046/j.1524-475X.1999.00335.x.
    1. Li J, Zhang YP, Kirsner RS. Angiogenesis in wound repair: angiogenic growth factors and the extracellular matrix. Microsc Res Tech. 2003;60:107–114. doi: 10.1002/jemt.10249.
    1. Carmeliet P, Moons L, Luttun A, Vincenti V, Compernolle V, De Mol M, Wu Y, Bono F, Devy L, Beck H, Scholz D, Acker T, DiPalma T, Dewerchin M, Noel A, Stalmans I, Barra A, Blacher S, VandenDriessche T, Ponten A, Eriksson U, Plate KH, Foidart JM, Schaper W, Charnock-Jones DS, Hicklin DJ, Herbert JM, Collen D, Persico MG. Synergism between vascular endothelial growth factor and placental growth factor contributes to angiogenesis and plasma extravasation in pathological conditions. Nat Med. 2001;7:575–583. doi: 10.1038/87904.
    1. Kim JC, Tseng SC. The effects on inhibition of corneal neovascularization after human amniotic membrane transplantation in severely damaged rabbit corneas. Korean J Ophthalmol. 1995;9:32–46.
    1. Bennett JP, Matthews R, Faulk WP. Treatment of chronic ulceration of the legs with human amnion. Lancet. 1980;1:1153–1156.
    1. Faulk WP, Matthews R, Stevens PJ, Bennett JP, Burgos H, Hsi BL. Human amnion as an adjunct in wound healing. Lancet. 1980;1:1156–1158.
    1. Hao Y, Ma DH, Hwang DG, Kim WS, Zhang F. Identification of antiangiogenic and antiinflammatory proteins in human amniotic membrane. Cornea. 2000;19:348–352. doi: 10.1097/00003226-200005000-00018.
    1. Steed DL, Trumpower C, Duffy D, Smith C, Marshall V, Rupp R, Robson M. Amnion-derived cellular cytokine solution: a physiological combination of cytokines for wound healing. Eplasty. 2008;8:e18.
    1. Uberti MG, Pierpont YN, Ko F, Wright TE, Smith CA, Cruse CW, Robson MC, Payne WG. Amnion-derived cellular cytokine solution (ACCS) promotes migration of keratinocytes and fibroblasts. Ann Plast Surg. 2010;64:632–635.

Source: PubMed

3
구독하다