Journal Home Online First Current Issue Archive For Authors Journal Information 中文版

Engineering >> 2021, Volume 7, Issue 2 doi: 10.1016/j.eng.2020.02.010

Enhancing the Surface Properties of a Bioengineered Anterior Cruciate Ligament Matrix for Use with Point-of-Care Stem Cell Therapy

a Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health, Farmington, CT 06030, USA
b Raymond and Beverly Sackler Center for Biological, Physical and Engineering Sciences, University of Connecticut Health, Farmington, CT 06030, USA
c Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT 06030, USA
d Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
e Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
f Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
g Department of Reconstructive Sciences, University of Connecticut Health, Farmington, CT 06030, USA
# These authors contributed equally to this work.

Received: 2019-08-08 Revised: 2019-12-26 Accepted: 2020-02-03 Available online: 2020-05-07

Next Previous

Abstract

We have previously developed a poly(L-lactic) acid (PLLA) bioengineered anterior cruciate ligament (ACL) matrix that has demonstrated enhanced healing when seeded with primary ACL cells prior to implantation in a rabbit model, as compared with the matrix alone. This suggests that improving cell adhesion on the matrix may beneficially affect the healing response and long-term performance of the bioengineered ACL matrix. One regenerative engineering approach involves enhancing the surface properties of the matrix to support cell adhesion and growth in combination with point-of-care stem cell therapy. Herein, we studied the cell adhesion properties of PLLA braided microfiber matrices enhanced through the physical adsorption of fibronectin and air plasma treatment. We evaluated the kinetics and binding efficiency of fibronectin onto matrices at three time points and three fibronectin concentrations. Incubating the matrix for 120 min in a solution of 25 μg·mL−1 fibronectin achieved the greatest binding efficiency to the matrix and cellular adhesion. Exposing the matrices to air plasma treatment for 5 min before fibronectin adsorption significantly enhanced the cell adhesion of rabbit bone marrow-derived mesenchymal stem cells (R-BMMSCs) 24 h post cell seeding. Finally, cellular proliferation was monitored for up to 21 d, the matrices were exposed to air plasma treatment, and fibronectin adsorption was found to result in enhanced cell number. These findings suggest that exposure to air plasma treatment and fibronectin adsorption enhances the cellular adhesion of PLLA braided microfiber matrices and may improve the clinical efficacy of the matrix in combination with point-of-care stem cell therapies.

Figures

Fig. 1

Fig. 2

Fig. 3

Fig. 4

Fig. 5

References

[ 1 ] Mather RC, Hettrich CM, Dunn WR, Cole BJ, Bach BR Jr, Huston LJ, et al. Costeffectiveness analysis of early reconstruction versus rehabilitation and delayed reconstruction for anterior cruciate ligament tears. Am J Sports Med 2014;42 (7):1583–91. link1

[ 2 ] Biau DJ, Tournoux C, Katsahian S, Schranz PJ, Nizard RS. Bone-patellar tendonbone autografts versus hamstring autografts for reconstruction of anterior cruciate ligament: meta-analysis. BMJ 2006;332(7548):995–1001. link1

[ 3 ] Predescu V, Georgeanu V, Prescura C, Stoian V, Cristea S. Anterior cruciate ligament reconstruction: soft tissue vs bone–tendon–bone. In: Proceedings of the 2nd Advanced Technologies for Enhanced Quality of Life; 2010 Jul 15–19; Iasi, Romania. New York: IEEE; 2010. p. 53–7.

[ 4 ] Magnussen RA, Carey JL, Spindler KP. Does autograft choice determine intermediate-term outcome of ACL reconstruction? Knee Surg Sports Traumatol Arthrosc 2011;19(3):462–72. link1

[ 5 ] Richmond JC, Manseau CJ, Patz R, McConville O. Anterior cruciate reconstruction using a Dacron ligament prosthesis. A long-term study. Am J Sports Med 1992;20(1):24–8. link1

[ 6 ] Moyen BJ, Jenny JY, Mandrino AH, Lerat JL. Comparison of reconstruction of the anterior cruciate ligament with and without a Kennedy ligamentaugmentation device. A randomized, prospective study. J Bone Joint Surg Am 1992;74(9):1313–9. link1

[ 7 ] Freeman JW, Kwansa AL. Recent advancements in ligament tissue engineering: the use of various techniques and materials for ACL repair. Recent Pat Biomed Eng 2008;1(1):18–23. link1

[ 8 ] Legnani C, Ventura A, Terzaghi C, Borgo E, Albisetti W. Anterior cruciate ligament reconstruction with synthetic grafts. A review of literature. Int Orthop 2010;34(4):465–71. link1

[ 9 ] Olson EJ, Kang JD, Fu FH, Georgescu HI, Mason GC, Evans CH. The biochemical and histological effects of artificial ligament wear particles: in vitro and in vivo studies. Am J Sports Med 1988;16(6):558–70. link1

[10] Mengsteab PY, Nair LS, Laurencin CT. The past, present and future of ligament regenerative engineering. Regen Med 2016;11:871–81. link1

[11] Mengsteab PY, McKenna M, Cheng J, Sun Z, Laurencin CT. Regenerative strategies for the treatment of knee joint disabilities. Berlin: Springer International Publishing; 2017. p. 391–410. link1

[12] Cooper JA, Sahota JS, Gorum WJ, Carter J, Doty SB, Laurencinl CT. Biomimetic tissue-engineered anterior cruciate ligament replacement. Proc Nat Acad Sci USA 2007;104(9):3049–54. link1

[13] Mengsteab PY, Conroy P, Badon M, Otsuka T, Kan HM, Vella AT, et al. Evaluation of a bioengineered ACL matrix’s osteointegration with BMP-2 supplementation. PLoS ONE 2020;15:1–18. link1

[14] Lu HH, Cooper JA, Manuel S, Freeman JW, Attawia MA, Ko FK, et al. Anterior cruciate ligament regeneration using braided biodegradable scaffolds: in vitro optimization studies. Biomaterials 2005;26:4805–16. link1

[15] Nair LS, Laurencin CT. Biodegradable polymers as biomaterials. Prog Polym Sci 2007;32:762–98. link1

[16] Laurencin CT, Freeman JW. Ligament tissue engineering: an evolutionary materials science approach. Biomaterials 2005;26:7530–6. link1

[17] Cooper JA, Lu HH, Ko FK, Freeman JW, Laurencin CT. Fiber-based tissueengineered scaffold for ligament replacement: design considerations and in vitro evaluation. Biomaterials 2005;26:1523–32. link1

[18] Yahia L. Ligaments and ligamentoplasties. Berlin: Springer; 1997. link1

[19] Cooper JA, Bailey LO, Carter JN, Castiglioni CE, Kofron MD, Ko FK, et al. Evaluation of the anterior cruciate ligament, medial collateral ligament, achilles tendon and patellar tendon as cell sources for tissue-engineered ligament. Biomaterials 2006;27:2747–54. link1

[20] Stevens MM, George JH. Exploring and engineering the cell surface interface. Science 2005;310(80):1135–8. link1

[21] Hallab NJ, Bundy KJ, O’Connor K, Moses RL, Jacobs JJ. Evaluation of metallic and polymeric biomaterial surface energy and surface roughness characteristics for directed cell adhesion. Tissue Eng 2001;7:55–71. link1

[22] Hanson AD, Wall ME, Pourdeyhimi B, Loboa EG. Effects of oxygen plasma treatment on adipose-derived human mesenchymal stem cell adherence to poly(L-lactic acid) scaffolds. J Biomater Sci Polym Ed 2007;18(11):1387–400. link1

[23] Rosso F, Giordano A, Barbarisi M, Barbarisi A. From cell-ECM interactions to tissue engineering. J Cell Physiol 2004;199(2):174–80. link1

[24] Sahoo S, Cho-Hong JG, Siew-Lok T. Development of hybrid polymer scaffolds for potential applications in ligament and tendon tissue engineering. Biomed Mater 2007;2:169–73. link1

[25] Leiss M, Beckmann K, Girós A, Costell M, Fässler R. The role of integrin binding sites in fibronectin matrix assembly in vivo. Curr Opin Cell Biol 2008;20:502–7. link1

[26] Millard CJ, Ellis IR, Pickford AR, Schor AM, Schor SL, Campbell ID. The role of the fibronectin IGD motif in stimulating fibroblast migration. J Biol Chem 2007;282(49):35530–5. link1

[27] Hall BK, Miyake T. All for one and one for all: condensations and the initiation of skeletal development. Bioassays 2000;22(2):138–47. link1

[28] Sechler JL, Schwarzbauer JE. Control of cell cycle progression by fibronectin matrix architecture. J Biol Chem 1998;273:25533–6. link1

[29] Cheng X, Gurkan UA, Dehen CJ, Tate MP, Hillhouse HW, Simpsonet GJ, et al. An electrochemical fabrication process for the assembly of anisotropically oriented collagen bundles. Biomaterials 2008;29:3278–88. link1

[30] Bhati RS, Mukherjee DP, McCarthy KJ, Rogers SH, Smith DF, Shalaby SW. The growth of chondrocytes into a fibronectin-coated biodegradable scaffold. J Biomed Mater Res 2001;56:74–82. link1

[31] García AJ, Vega MD, Boettiger D. Modulation of cell proliferation and differentiation through substrate-dependent changes in fibronectin conformation. Mol Biol Cell 1999;10:785–98. link1

[32] Gelberman RH, Steinberg D, Amiel D, Akeson W. Fibroblast chemotaxis after tendon repair. J Hand Surg Am 1991;16:686–93. link1

[33] Neurath M. Expression of tenascin, laminin and fibronectin following traumatic rupture of the anterior cruciate ligament. Z Orthop Ihre Grenzgeb 1993;131(2):168–72. link1

[34] Amiel D, Foulk RA, Harwood FL, Akeson WH. Quantitative assessment by competitive ELISA of fibronectin (Fn) in tendons and ligaments. Matrix 1990;9:421–7. link1

[35] Caplan AI. Adult mesenchymal stem cells for tissue engineering versus regenerative medicine. J Cell Physiol 2007;213:341–7. link1

[36] Caterson EJ, Nesti LJ, Danielson KG, Tuan RS. Human marrow-derived mesenchymal progenitor cells. Mol Biotechnol 2002;20:245–56. link1

[37] Jiang Y, Jahagirdar BN, Reinhardt RE, Schwartz RE, Keene CD, Ortiz-Gonzalezet XR, et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 2002;418(6893):41–9. link1

[38] Jager M, Jelinek EM, Wess KM, Scharfstadt A, Jacobson M, Kevy SV, et al. Bone marrow concentrate: a novel strategy for bone defect treatment. Curr Stem Cell Res Ther 2009;4(1):34–43. link1

[39] Segers VFM, Lee RT. Stem-cell therapy for cardiac disease. Nature 2008;451:937–42. link1

[40] Gómez-Barrena E, Rosset P, Müller I, Giordano R, Bunu C, Layrolle P, et al. Bone regeneration: stem cell therapies and clinical studies in orthopaedics and traumatology. J Cell Mol Med 2011;15:1266–86. link1

[41] Jäger M, Herten M, Fochtmann U, Fischer J, Hernigou P, Zilkens C, et al. Bridging the gap: bone marrow aspiration concentrate reduces autologous bone grafting in osseous defects. J Orthop Res 2011;29:173–80. link1

[42] Roh JD, Nelson GN, Udelsman BV, Brennan MP, Lockhart B, Fong PM, et al. Centrifugal seeding increases seeding efficiency and cellular distribution of bone marrow stromal cells in porous biodegradable scaffolds. Tissue Eng 2007;13:2743–9. link1

[43] Hisatome T, Yasunaga Y, Yanada S, Tabata Y, Ikada Y, Ochi M. Neovascularization and bone regeneration by implantation of autologous bone marrow mononuclear cells. Biomaterials 2005;26(22):4550–6. link1

[44] Hibino N, Nalbandian A, Devine L, Martinez RS, McGillicuddy E, Yi T, et al. Comparison of human bone marrow mononuclear cell isolation methods for creating tissue-engineered vascular grafts: novel filter system versus traditional density centrifugation method. Tissue Eng Part C Methods 2011;17:993–8. link1

[45] Neen D, Noyes D, Shaw M, Gwilym S, Fairlie N, Birch N. Healos and bone marrow aspirate used for lumbar spine fusion. Spine 2006;31(18):E636–40. link1

[46] Hindié M, Degat MC, Gaudière F, Gallet O, Van Tassel PR, Pauthe E. Preosteoblasts on poly(L-lactic acid) and silicon oxide: Influence of fibronectin and albumin adsorption. Acta Biomater 2011;7(1):387–94. link1

[47] Xu H, Deshmukh R, Timmons R, Nguyen KT. Enhanced endothelialization on surface modified poly(L-lactic acid) substrates. Tissue Eng Part A 2011;17:865–76. link1

[48] Jiao Y, Zhou C, Li L. Protein adsorption on the poly(L-lactic acid) surface modified by chitosan and its derivatives. Chinese Sci Bull 2009;54 (18):3167–73. link1

[49] Teraoka F, Nakagawa M, Hara M. Surface modification of poly(L-lactide) by atmospheric pressure plasma treatment and cell response. Dent Mater J 2006;25:560–5. link1

[50] Nakagawa M, Teraoka F, Fujimoto S, Hamada Y, Kibayashi H, Takahashi J. Improvement of cell adhesion on poly(L-lactide) by atmospheric plasma treatment. J Biomed Mater Res 2006;77(1):112–8. link1

[51] Deligianni DD, Katsala ND, Koutsoukos PG, Missirlis YF. Effect of surface roughness of hydroxyapatite on human bone marrow cell adhesion, proliferation, differentiation and detachment strength. Biomaterials 2000;22:87–96. link1

[52] Schlie-Wolter S, Ngezahayo A, Chichkov BN. The selective role of ECM components on cell adhesion, morphology, proliferation and communication in vitro. Exp Cell Res 2013;319:1553–61. link1

[53] Keselowsky BG, Collard DM, García AJ. Surface chemistry modulates fibronectin conformation and directs integrin binding and specificity to control cell adhesion. J Biomed Mater Res 2003;66A(Pt A):247–59. link1

[54] Jeon H, Simon CG, Kim G. A mini-review: cell response to microscale, nanoscale, and hierarchical patterning of surface structure. J Biomed Mater Res Part B Appl Biomater 2014;102(7):1580–94. link1

[55] Bettinger CJ, Langer R, Borenstein JT. Engineering substrate topography at the micro- and nanoscale to control cell function. Angew Chem Int Ed 2009;48:5406–15. link1

[56] Figueroa D, Calvo R, Vaisman A, Arellano S, Figueroa F, Donoso R, et al. Arthroscopic intercondylar notch bone marrow aspiration during anterior cruciate ligament reconstruction. Arthrosc Tech 2019;8(12): e1437–41. link1

[57] Truskey GA, Proulx TL. Relationship between 3T3 cell spreading and the strength of adhesion on glass and silane surfaces. Biomaterials 1993;14 (4):243–54. link1

Related Research