<iframe src="//www.googletagmanager.com/ns.html?id=GTM-5TSRKG" height="0" width="0" style="display: none; visibility: hidden">
Review Article
No access
Published Online: 12 January 2016

Topical Collagen-Based Biomaterials for Chronic Wounds: Rationale and Clinical Application

Publication: Advances in Wound Care
Volume 5, Issue Number 1

Abstract

Significance: The extracellular matrix (ECM) is known to be deficient in chronic wounds. Collagen is the major protein in the ECM. Many claims are made while extolling the virtues of collagen-based biomaterials in promoting cell growth and modulating matrix metalloproteinases. This review will explore the rationale for using topical collagen or ECM as an interface for healing.
Recent Advances: Rapid improvements in electrospinning and nanotechnology have resulted in the creation of third-generation biomaterials that mimic the native ECM, stimulate cellular and genetic responses in the target tissue, and provide a platform for controlled release of bioactive molecules and live cells. Although the major focus is currently on development of artificial tissues and organ regeneration, better understanding of the mechanisms that stimulate wound healing can be applied to specific deficits in the chronic wound.
Critical Issues: When choosing between the various advanced wound-care products and dressings, the clinician is challenged to select the most appropriate material at the right time. Understanding how the ECM components promote tissue regeneration and modulate the wound microenvironment will facilitate those choices. Laboratory discoveries of biomolecular and cellular strategies that promote skin regeneration rather than repair should be demonstrated to translate to deficits in the chronic wound.
Future Directions: Cost-effective production of materials that utilize non-mammalian sources of collagen or ECM components combined with synthetic scaffolding will provide an optimal structure for cellular ingrowth and modulation of the chronic wound microenvironment to facilitate healing. These bioengineered materials will be customizable to provide time-released delivery of bioactive molecules or drugs based on the degradation rate of the scaffold or specific signals from the wound.

Get full access to this article

View all available purchase options and get full access to this article.

About the Author

Lisa J. Gould, MD, PhD, is the Medical Director of the Kent Hospital Wound Recovery and Hyperbaric Medicine Center and an Affiliate Professor in the Department of Molecular Pharmacology and Physiology at the University of South Florida. Her clinical and basic science research focuses on ischemic and chronic wounds in older adults.

References

1.
Fife CE, Carter MJ, Walker D, Thomson B. Wound care outcomes and associated cost among patients treated in US outpatient wound centers: data from the US wound registry. Wounds 2012;24:10–17.
2.
Schultz GS, Wysocki A. Interactions between extracellular matrix and growth factors in wound healing. Wound Repair Regen 2009;17:153–162.
3.
Black E, et al. Decrease of collagen deposition in wound repair in type 1 diabetes independent of glycemic control. Arch Surg 2003;138:34–40.
4.
Cullen B, Smith R, Mcculloch E, Silcock D, Morrison L. Mechanism of action of PROMOGRAN, a protease modulating matrix, for the treatment of diabetic foot ulcers. Wound Repair Regen 2002;10:16–25.
5.
Bermudez DM, et al. Impaired biomechanical properties of diabetic skin. Am J Pathol 2011;178:2215–2223.
6.
Lerman OZ, Galiano RD, Armour M, Levine JP, Gurtner GC. Cellular dysfunction in the diabetic fibroblast: impairment in migration, vascular endothelial growth factor production, and response to hypoxia. Am J Pathol 2003;162:303–312.
7.
Loot MAM, et al. Fibroblasts derived from chronic diabetic ulcers differ in their response to stimulation with EGF, IGF-I, bFGF and PDGF-AB compared to controls. Eur J Cell Biol 2002;81:153–160.
8.
Cook H, Stephens P, Davies KJ, Harding KG, Thomas DW. Defective extracellular matrix reorganization by chronic wound fibroblasts is associated with alterations in TIMP-1, TIMP-2, and MMP-2 activity. J Invest Dermatol 2000;115:225–233.
9.
Pastar I, et al. Epithelialization in wound healing: a comprehensive review. Adv Wound Care 2014;3:445–464.
10.
Volk SW, Iqbal SA, Bayat A. Interactions of the extracellular matrix and progenitor cells in cutaneous wound healing. Adv Wound Care 2013;2:261–272.
11.
Stojadinovic O, et al. Deregulation of keratinocyte differentiation and activation: a hallmark of venous ulcers. J Cell Mol Med 2008;12:2675–2690.
12.
Usui ML, Mansbridge JN, Carter WG, Fujita M, Olerud JE. Keratinocyte migration, proliferation, and differentiation in chronic ulcers from patients with diabetes and normal wounds. J Histochem Cytochem 2008;56:687–696.
13.
Shoulders MD, Raines RT. Collagen structure and stability. Annu Rev Biochem 2009;78:929–958.
14.
Gautam S, Chou C-F, Dinda AK, Potdar PD, Mishra NC. Surface modification of nanofibrous polycaprolactone/gelatin composite scaffold by collagen type I grafting for skin tissue engineering. Mater Sci Eng C 2014;34:402–409.
15.
Parenteau-Bareil R, et al. Comparative study of bovine, porcine and avian collagens for the production of a tissue engineered dermis. Acta Biomater 2011;7:3757–3765.
16.
Neve A, Cantatore FP, Maruotti N, Corrado A, Ribatti D. Extracellular matrix modulates angiogenesis in physiological and pathological conditions. BioMed Res Int 2014;2014:1–10.
17.
Whelan MC. Collagen I initiates endothelial cell morphogenesis by inducing actin polymerization through suppression of cyclic AMP and protein kinase A. J Biol Chem 2002;278:327–334.
18.
Wiegand C, et al. Protease and pro-inflammatory cytokine concentrations are elevated in chronic compared to acute wounds and can be modulated by collagen type I in vitro. Arch Dermatol Res 2010;302:419–428.
19.
Wiegand C, et al. Effect of the sterilization method on the performance of collagen type I on chronic wound parameters in vitro. J Biomed Mater Res B Appl Biomater 2009;90B:710–719.
20.
Schönfelder U, et al. Influence of selected wound dressings on PMN elastase in chronic wound fluid and their antioxidative potential in vitro. Biomaterials 2005;26:6664–6673.
21.
Friess W. Collagen—biomaterial for drug delivery. Eur J Pharm 1998;45:113–136.
22.
Goo HC, Hwang Y-S, Choi YR, Cho HN, Suh H. Development of collagenase-resistant collagen and its interaction with adult human dermal fibroblasts. Biomaterials 2003;24:5099–5113.
23.
Chattopadhyay S, Raines RT. Review collagen-based biomaterials for wound healing: collagen-Based Biomaterials. Biopolymers 2014;101:821–833.
24.
Boyce ST, Warden GD. Principles and practices for treatment of cutaneous wounds with cultured skin substitutes. Am J Surg 2002;183:445–456.
25.
Harriger MD, Supp AP, Warden GD, Boyce ST. Glutaraldehyde crosslinking of collagen substrates inhibits degradation in skin substitutes grafted to athymic mice. J Biomed Mater Res 1997;35:137–145.
26.
Cuttle L, et al. Collagen in the scarless fetal skin wound: Detection with Picrosirius-polarization. Wound Repair Regen 2005;13:198–204.
27.
Rolfe KJ, Grobbelaar AO. A review of fetal scarless healing. ISRN Dermatol 2012;2012:1–9.
28.
Zgheib C, Xu J, Liechty KW. Targeting inflammatory cytokines and extracellular matrix composition to promote wound regeneration. Adv Wound Care 2014;3:344–355.
29.
Larson BJ, Longaker MT, Lorenz HP. Scarless fetal wound healing: a basic science review. Plast Reconstr Surg 2010;126:1172–1180.
30.
Liu X, Wu H, Byrne M, Krane S, Jaenisch R. Type III collagen is crucial for collagen I fibrillogenesis and for normal cardiovascular development. Proc Natl Acad Sci U S A 1997;94:1852–1856.
31.
Volk SW, Wang Y, Mauldin EA, Liechty KW, Adams SL. Diminished type III collagen promotes myofibroblast differentiation and increases scar deposition in cutaneous wound healing. Cells Tissues Organs 2011;194:25–37.
32.
Nuutila K, et al. Recombinant human collagen III gel for transplantation of autologous skin cells in porcine full-thickness wounds: rhCol-III gel for wound healing. J Tissue Eng Regen Med [Epub ahead of print];
33.
KüHn K, et al. The structure of type IV collagen. Ann N Y Acad Sci 1985;460:14–24.
34.
Abreu-Velez A, Howard M. Collagen IV in normal skin and in pathological processes. North Am J Med Sci 2012;4:1.
35.
Ortega N. New functional roles for non-collagenous domains of basement membrane collagens. J Cell Sci 2002;115:4201–4214.
36.
Viglio S, et al. Wound repair capability in EDS fibroblasts can be retrieved by exogenous type V collagen. Sci World J 2008;8:956–958.
37.
Viglio S, et al. Rescue of migratory defects of Ehlers–Danlos syndrome fibroblasts in vitro by type v collagen but not insulin-like binding protein-1. J Invest Dermatol 2008;128:1915–1919.
38.
Inkinen K, Turakainen H, Wolff H, Ahonen J. Cloning of cDNA for Rat Pro α1(V) Collagen mRNA. Expression patterns of type I, type III and type V collagen genes in experimental granulation tissue. Connect Tissue Res 1999;40:209–220.
39.
Inkinen K, Wolff H, Von Boguslawski K, Ahonen J. Type V collagen in experimental granulation tissue. Connect Tissue Res 1998;39:281–294.
40.
Bruckner-Tuderman L. Dystrophic epidermolysis bullosa: pathogenesis and clinical features. Dermatol Clin 2010;28:107–114.
41.
Nyström A, et al. Collagen VII plays a dual role in wound healing. J Clin Invest 2013;123:3498–3509.
42.
Wang X, et al. Topical application of recombinant type VII collagen incorporates into the dermal–epidermal junction and promotes wound closure. Mol Ther 2013;21:1335–1344.
43.
Ruszczak Z. Effect of collagen matrices on dermal wound healing. Adv Drug Deliv Rev 2003;55:1595–1611.
44.
Westgate S, Cutting K, DeLuca G, Asaad K. Collagen dressings made easy. Wounds UK 2012;8.
45.
Peng YY, et al. Towards scalable production of a collagen-like protein from Streptococcus pyogenes for biomedical applications. Microb Cell Factories 2012;11:146.
46.
Shilo S, et al. Cutaneous wound healing after treatment with plant-derived human recombinant collagen flowable gel. Tissue Eng Part A 2013;19:1519–1526.
47.
Lun S, et al. A functional extracellular matrix biomaterial derived from ovine forestomach. Biomaterials 2010;31:4517–4529.
48.
Youngstrom DW, Barrett JG, Jose RR, Kaplan DL. Functional characterization of detergent-decellularized equine tendon extracellular matrix for tissue engineering applications. PLoS One 2013;8:e64151.
49.
Badylak SF. The extracellular matrix as a biologic scaffold material. Biomaterials 2007;28:3587–3593.
50.
Lin H-K, et al. Understanding roles of porcine small intestinal submucosa in urinary bladder regeneration: identification of variable regenerative characteristics of small intestinal submucosa. Tissue Eng Part B Rev 2014;20:73–83.
51.
Brennan EP, et al. Antibacterial activity within degradation products of biological scaffolds composed of extracellular matrix. Tissue Eng 2006;12:2949–2955.
52.
Brennan EP, Tang X-H, Stewart-Akers AM, Gudas LJ, Badylak SF. Chemoattractant activity of degradation products of fetal and adult skin extracellular matrix for keratinocyte progenitor cells. J Tissue Eng Regen Med 2008;2:491–498.
53.
Sarikaya A, et al. Antimicrobial activity associated with extracellular matrices. Tissue Eng 2002;8:63–71.
54.
Li F, et al. Low-molecular-weight peptides derived from extracellular matrix as chemoattractants for primary endothelial cells. Endothelium 2004;11:199–206.
55.
Carey LE, et al. In vivo degradation of 14C-labeled porcine dermis biologic scaffold. Biomaterials 2014;35:8297–8304.
56.
Shi L, Ronfard V. Biochemical and biomechanical characterization of porcine small intestinal submucosa (SIS): a mini review. Int J Burns Trauma 2013;3:173–179.
57.
Sicari BM, et al. The effect of source animal age upon the in vivo remodeling characteristics of an extracellular matrix scaffold. Biomaterials 2012;33:5524–5533.
58.
Tottey S, et al. The effect of source animal age upon extracellular matrix scaffold properties. Biomaterials 2011;32:128–136.
59.
Crapo PM, Gilbert TW, Badylak SF. An overview of tissue and whole organ decellularization processes. Biomaterials 2011;32:3233–3243.
60.
You C, Wang X, Zheng Y, Han C. Three types of dermal grafts in rats: the importance of mechanical property and structural design. Biomed Eng OnLine 2013;12:125.
61.
Greer N, et al. Advanced wound care therapies for nonhealing diabetic, venous, and arterial ulcers: a systematic review. Ann Intern Med 2013;159:532.
62.
Holmes C, Wrobel J, Mac Eachern MP, Boles BR. Collagen-based wound dressings for the treatment of diabetes-related foot ulcers: a systematic review. Diabetes Metab Syndr Obes Targets Ther 2013;6:17–29.
63.
Valle MF, et al. Comparative effectiveness of advanced wound dressings for patients with chronic venous leg ulcers: a systematic review. Wound Repair Regen 2014;22:193–204.
64.
Harding KG, Kirsner RS, Lee D, Mulder G, Serena T. International consensus. Acellular matrices for the treatment of wounds. An expert working group review. Wounds Int 2010. www.woundsinternational.com/pdf/content_9732.pdf (last accessed October 11, 2014).
65.
Snyder DL, Sullivan N, Schoelles KM. Skin substitutes for treating chronic wounds technology assessment report. (2012). www.ahrq.gov/research/findings/ta/skinsubs/HCPRO610-skinsubst-final.pdf (last accessed October 11, 2014).
66.
Hankin CS, Knispel J, Lopes M, Bronstone A, Maus E. Clinical and cost efficacy of advanced wound care matrices for venous ulcers. J Manag Care Pharm 2012;18:375–384.
67.
O'Donnell TF, Lau J. A systematic review of randomized controlled trials of wound dressings for chronic venous ulcer. J Vasc Surg 2006;44:1118–1125.
68.
Lev-Tov H, Li C-S, Dahle S, Isseroff RR. Cellular versus acellular matrix devices in treatment of diabetic foot ulcers: study protocol for a comparative efficacy randomized controlled trial. Trials 2013;14:8.
69.
Dimitrijevich SD, et al. In vivo degradation of oxidized, regenerated cellulose. Carbohydr Res 1990;198:331–341.
70.
Hart J, et al. The role of oxidised regenerated cellulose/collagen in wound repair: effects in vitro on fibroblast biology and in vivo in a model of compromised healing. Int J Biochem Cell Biol 2002;34:1557–1570.
71.
Cullen B, et al. The role of oxidised regenerated cellulose/collagen in chronic wound repair and its potential mechanism of action. Int J Biochem Cell Biol 2002;34:1544–1556.
72.
Pierce AM, Wiebkin OW, Wilson DF. Surgicel: its fate following implantation. J Oral Pathol 1984;13:661–670.
73.
Gottrup F, et al. Randomized controlled trial on collagen/oxidized regenerated cellulose/silver treatment: RCT on collagen/ORC/silver in diabetic foot ulcers. Wound Repair Regen 2013;21:216–225.
74.
Ulrich D, Smeets R, Unglaub F, Wöltje M, Pallua N. Effect of oxidized regenerated cellulose/collagen matrix on proteases in wound exudate of patients with diabetic foot ulcers. J Wound Ostomy Continence Nurs 2011;38:522–528.
75.
McCarty SM, Percival SL. Proteases and delayed wound healing. Adv Wound Care 2013;2:438–447.
76.
Hoganson DM, et al. The retention of extracellular matrix proteins and angiogenic and mitogenic cytokines in a decellularized porcine dermis. Biomaterials 2010;31:6730–6737.
77.
Sicari BM, et al. An acellular biologic scaffold promotes skeletal muscle formation in mice and humans with volumetric muscle loss. Sci Transl Med 2014;6:234ra58.
78.
Pashuck ET, Stevens MM. Designing regenerative biomaterial therapies for the clinic. Sci Transl Med 2012;4:160sr4.
79.
Muthukumar T, Prabu P, Ghosh K, Sastry TP. Fish scale collagen sponge incorporated with Macrotyloma uniflorum plant extract as a possible wound/burn dressing material. Colloids Surf B Biointerfaces 2014;113:207–212.
80.
Stankus JJ, Freytes DO, Badylak SF, Wagner WR. Hybrid nanofibrous scaffolds from electrospinning of a synthetic biodegradable elastomer and urinary bladder matrix. J Biomater Sci Polym Ed 2008;19:635–652.
81.
Jayarama Reddy V, et al. Nanofibrous structured biomimetic strategies for skin tissue regeneration: nanofibrous structures for wound healing. Wound Repair Regen 2013;21:1–16.
82.
Gibson M, et al. Tissue extracellular matrix nanoparticle presentation in electrospun nanofibers. BioMed Res Int 2014;2014:1–13.
83.
Bhat S, Kumar A. Biomaterials and bioengineering tomorrow's healthcare. Biomatter 2013;3:e24717.
84.
Machula H, Ensley B, Kellar R. Electrospun tropoelastin for delivery of therapeutic adipose-derived stem cells to full-thickness dermal wounds. Adv Wound Care 2014;3:367–375.
85.
Ricard-Blum S, Salza R. Matricryptins and matrikines: biologically active fragments of the extracellular matrix. Exp Dermatol 2014;23:457–463.
86.
Fang M, et al. Type I collagen D-spacing in fibril bundles of dermis, tendon, and bone: bridging between nano- and micro-level tissue hierarchy. ACS Nano 2012;6:9503–9514.
87.
Kalluri R. Angiogenesis: Basement membranes: structure, assembly and role in tumour angiogenesis. Nat Rev Cancer 2003;3:422–433.
88.
Oliveira AC, et al. Evaluation of small intestine grafts decellularization methods for corneal tissue engineering. PLoS One 2013;8:e66538.
89.
DiDomenico L, Emch KJ, Landsman AR, Landsman AA. Prospective comparison of diabetic foot ulcers treated with either a cryopreserved skin allograft or a bioengineered skin substitute. Wounds 2011;23:184–189.
90.
Reyzelman A, et al. Clinical effectiveness of an acellular dermal regenerative tissue matrix compared to standard wound management in healing diabetic foot ulcers: a prospective, randomised, multicentre study. J Int Wound 2009;6:196–208.
91.
Brigido SA. The use of an acellular dermal regenerative tissue matrix in the treatment of lower extremity wounds: a prospective 16-week pilot study. J Int Wound 2006;3:181–187.
92.
Brigido SA, Boc SF, Lopez RC. Effective management of major lower extremity wounds using an acellular regenerative tissue matrix: a pilot study. Orthopedics 2004;27:145–149.
93.
Romanelli M, Dini V, Bertone MS. Randomized comparison of OASIS wound matrix versus moist wound dressing in the treatment of difficult-to-heal wounds of mixed arterial/venous etiology. Adv Skin Wound Care 2010;23:34–38.
94.
Romanelli M, Dini V, Bertone M, Barbanera S, Brilli C. OASIS wound matrix versus hyaloskin in the treatment of difficult-to-heal wounds of mixed arterial/venous aetiology. J Int Wound 2007;4:3–7.
95.
Mostow EN, Haraway GD, Dalsing M, Hodde JP, King D. Effectiveness of an extracellular matrix graft (OASIS Wound Matrix) in the treatment of chronic leg ulcers: a randomized clinical trial. J Vasc Surg 2005;41:837–843.
96.
Niezgoda JA, Van Gils CC, Frykberg RG, Hodde JP. Randomized clinical trial comparing OASIS wound matrix to regranex gel for diabetic ulcers. Adv Skin Wound Care 2005;18:258–266.
97.
Gottrup F, et al. Randomized controlled trial on collagen/oxidized regenerated cellulose/silver treatment: RCT on collagen/ORC/silver in diabetic foot ulcers. Wound Repair Regen 2013;21:216–225.
98.
Veves A, Sheehan P, Pham HT. A randomized, controlled trial of Promogran (a collagen/oxidized regenerated cellulose dressing) vs standard treatment in the management of diabetic foot ulcers. Arch Surg 2002;137:822–827.
99.
Vin F, Teot L, Meaume S. The healing properties of Promogran in venous leg ulcers. J Wound Care 2002;11:335–341.
100.
Donaghue VM, et al. Evaluation of a collagen-alginate wound dressing in the management of diabetic foot ulcers. Adv Wound Care 1998;11:114–119.

Information & Authors

Information

Published In

cover image Advances in Wound Care
Advances in Wound Care
Volume 5Issue Number 1January 2016
Pages: 19 - 31

History

Published online: 12 January 2016
Published in print: January 2016
Published ahead of print: 5 January 2015
Accepted: 31 October 2014
Received: 21 August 2014

Permissions

Request permissions for this article.

Topics

Authors

Affiliations

Lisa J. Gould* [email protected]
Wound Recovery and Hyperbaric Medicine Center, Kent Hospital, Warwick, Rhode Island.

Notes

*
Correspondence: Wound Recovery and Hyperbaric Medicine Center, Kent Hospital, 455 Tollgate Rd, Warwick, RI 02886 (e-mail: [email protected]).

Author Disclosure and Ghostwriting

No competing financial interests exist. The content of this article was expressly written by the author. No ghostwriters were used to write this article.

Metrics & Citations

Metrics

Citations

Export citation

Select the format you want to export the citations of this publication.

View Options

Get Access

Access content

To read the fulltext, please use one of the options below to sign in or purchase access.

Society Access

If you are a member of a society that has access to this content please log in via your society website and then return to this publication.

Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

View options

PDF/EPUB

View PDF/ePub

Full Text

View Full Text

Media

Figures

Other

Tables

Share

Share

Copy the content Link

Share on social media

Back to Top