We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Heparin-releasing scaffold for stem cells: a differentiating device for vascular aims

    Cristiano Spadaccio

    Area of Cardiovascular Surgery, Center of Integrated Research, University Campus Bio-Medico of Rome, Italy

    Cardiac & Molecular Biology Laboratory, Heart, Lung & Esophageal Surgery Institute University of Pittsburgh Medical Center, PA, USA; McGowan Institute for Regenerative Medicine, PA, USA

    ,
    Alberto Rainer

    Laboratory of Chemistry & Biomaterials, Center of Integrated Research, Italy

    ,
    Matteo Centola

    Laboratory of Chemistry & Biomaterials, Center of Integrated Research, Italy

    ,
    Marcella Trombetta

    Laboratory of Chemistry & Biomaterials, Center of Integrated Research, Italy

    ,
    Massimo Chello

    Area of Cardiovascular Surgery, Center of Integrated Research, University Campus Bio-Medico of Rome, Italy

    ,
    Mario Lusini

    Area of Cardiovascular Surgery, Center of Integrated Research, University Campus Bio-Medico of Rome, Italy

    ,
    Elvio Covino

    Area of Cardiovascular Surgery, Center of Integrated Research, University Campus Bio-Medico of Rome, Italy

    ,
    Yoshiya Toyoda

    Cardiac & Molecular Biology Laboratory, Heart, Lung & Esophageal Surgery Institute University of Pittsburgh Medical Center, PA, USA; McGowan Institute for Regenerative Medicine, PA, USA

    &
    Published Online:https://doi.org/10.2217/rme.10.25

    Aims: Current limitations of tissue-engineered vascular grafts include timing for the scaffold preparation, cell type, cell differentiation and growth inside the construct, and thrombogenicity of the final device. To surmount these shortcomings, we developed a heparin-releasing poly-L-lactide (PLLA) scaffold using the electrospinning technique, to guide the differentiation of human mesenchymal stem cells towards the endothelial phenotype and to deliver a useful drug in the management of the postimplantation period. Materials & methods: The heparin-releasing PLLA scaffold was produced by means of the electrospinning technique in a tubular shape. The scaffold was seeded with human mesenchymal stem cells and cultured for up to 1 week. Cell viability and cytotoxicity assays were performed, and cell differentiation was evaluated by immunofluorescence with confocal microscopy, cytofluorometry and western blotting. Heparin release was assayed by Azure A method and biological effectiveness of the drug was assessed by activated clotting time measurements. Results: The scaffold exhibited a morphology favorable to cell attachment. Heparin release showed an initial burst within the first 24 h, followed by a further sustained release profile. After 48 h of culturing, the construct demonstrated adequate engraftment and viability. Increased proliferation compared with the control scaffold in bare PLLA, suggested the induction of a favorable microenvironment. A shift towards CD31 positivity and modifications in cell morphology were observed in the heparin-releasing PLLA scaffold. Conclusion: By exploiting the biological effects of heparin, we developed an ad hoc differentiating device towards the endothelial phenotype for autologous stem cell seeding and, at the same time, we were able to facilitate and optimize the management of the construct once in clinical settings.

    Papers of special note have been highlighted as: ▪ of interest

    Bibliography

    • Shin’oka T, Shum-Tim D, Ma PX et al.: Creation of viable pulmonary artery autografts through tissue engineering. J. Thorac. Cardiovasc. Surg.115(3),536–545; discussion 545–536 (1998).
    • Shin’oka T, Matsumura G, Hibino N et al.: Midterm clinical result of tissue-engineered vascular autografts seeded with autologous bone marrow cells. J. Thorac. Cardiovasc. Surg.129(6),1330–1338 (2005).
    • Sarkar S, Lee GY, Wong JY, Desai TA: Development and characterization of a porous micro-patterned scaffold for vascular tissue engineering applications. Biomaterials27(27),4775–4782 (2006).
    • Greisler HP, Cziperle DJ, Kim DU et al.: Enhanced endothelialization of expanded polytetrafluoroethylene grafts by fibroblast growth factor type 1 pretreatment. Surgery112(2),244–254; discussion 254–245 (1992).
    • Spadaccio C, Chello M, Trombetta M, Rainer A, Toyoda Y, Genovese J: Drug releasing systems in cardiovascular tissue engineering. J. Cell. Mol. Med.13(3),422–439 (2009).▪ Provides an extensive overview on tissue engineering for cardiovascular structures, with particular attention to scaffold fabrication techniques, functionalization with bioactive molecules, drug delivery systems and their applications in this field.
    • Xu C, Inai R, Kotaki M, Ramakrishna S: Electrospun nanofiber fabrication as synthetic extracellular matrix and its potential for vascular tissue engineering. Tissue Eng.10(7–8),1160–1168 (2004).▪ Provides an extensive description of the characteristics and advantages of nanofabrication by means of the electrospinning technique, underlining the importance of mimicking extracellular matrix structural properties in vascular tissue engineering.
    • Badami AS, Kreke MR, Thompson MS, Riffle JS, Goldstein AS: Effect of fiber diameter on spreading, proliferation, and differentiation of osteoblastic cells on electrospun poly(lactic acid) substrates. Biomaterials27(4),596–606 (2006).
    • Hashi CK, Zhu Y, Yang GY et al.: Antithrombogenic property of bone marrow mesenchymal stem cells in nanofibrous vascular grafts. Proc. Natl Acad. Sci. USA104(29),11915–11920 (2007).▪ Experimental work confirms the advantage of mimicking the arrangement of connective tissue fibrillar proteins using nano- and microfibrous electrospun scaffolds. Also underlines the role of mesenchymal stem cells as a potential alternative to autologous endothelial cells in a tissue-engineered vascular graft, for their easy availability and recruitment, high plasticity and antithrombogenic properties probably related to the presence of heparin-like proteoglicans expressed on their membrane.
    • Luong-Van E, Grondahl L, Chua KN, Leong KW, Nurcombe V, Cool SM: Controlled release of heparin from poly(e-caprolactone) electrospun fibers. Biomaterials27(9),2042–2050 (2006).▪ Represents an example of successful polymer functionalization performed on electrospun poly-caprolactone nanofibers, providing important data on heparin-release assays and interesting references on electrospun fibers drug loading.
    • 10  Huang ZM, Zhang ZY, Kotaki M, Ramakrishna S: A review on polymer nanofibers by electrospinning and their applications in nanocomposites. Compos. Sci. Technol.63,2223–2253 (2003).
    • 11  Salacinski HJ, Tiwari A, Hamilton G, Seifalian AM: Cellular engineering of vascular bypass grafts: role of chemical coatings for enhancing endothelial cell attachment. Med. Biol. Eng. Comput.39(6),609–618 (2001).▪ Describes current limitations of tissue-engineered vascular grafts mainly concerning cell type, availability, differentiation and growth inside the construct, thrombogenicity, mechanical resistance, and reasonable and clinically suitable timing for scaffold preparation.
    • 12  Wight TN: Cell biology of arterial proteoglycans. Arteriosclerosis9(1),1–20 (1989).
    • 13  Zilla P, Deutsch M, Meinhart J et al.: Clinical in vitro endothelialization of femoropopliteal bypass grafts: an actuarial follow-up over three years. J. Vasc. Surg.19(3),540–548 (1994).
    • 14  Rosenman JE, Kempczinski RF, Pearce WH, Silberstein EB: Kinetics of endothelial cell seeding. J. Vasc. Surg.2(6),778–784 (1985).
    • 15  Spadaccio C, Rainer A, Chello M, Covino E, Trombetta M, Genovese J: Drug releasing hybrid scaffold: new avenue in cardiovascular tissue engineering. Tissue Eng. Part A14(5),691–943 (2008).▪ Contains preliminary data on the use of heparin functionalized electrospun scaffolds to produce tailored extracellular matrix (ECM)-like differentiating devices using human mesenchymal stem cells for cardiovascular applications.
    • 16  Spadaccio C, Rainer A, Trombetta M et al.: Poly-L-lactic acid/hydroxyapatite electrospun nanocomposites induce chondrogenic differentiation of human MSC. Ann. Biomed. Eng.37(7),1376–1389 (2009).▪ Represents a proof of principle of the possibility to produce a scaffold suitable for stem cell seeding, containing the appropriate factors to induce a guided differentiation towards the desired phenotype within a 3D ECM-like environment closely mimicking the native tissue histoarchitecture, and allowing a harmonious ongoing growth and differentiation for tissue regeneration.
    • 17  Lobo BL, Greene WL: ACCP consensus conference on antithrombotic therapy. Indicate specific low molecular weight heparin product and dosage. Chest110(3),866 (1996).
    • 18  Rawat A, Yang T, Hussain A, Ahsan F: Complexation of a poly-L-arginine with low molecular weight heparin enhances pulmonary absorption of the drug. Pharm. Res.25(4),936–948 (2008).
    • 19  Sell SA, McClure MJ, Barnes CP et al.: Electrospun polydioxanone-elastin blends: potential for bioresorbable vascular grafts. Biomed. Mater.1(2),72–80 (2006).
    • 20  Smith MJ, Mcclure MJ, Sell SA et al.: Suture-reinforced electrospun polydioxanone-elastin small-diameter tubes for use in vascular tissue engineering: a feasibility study. Acta Biomater.4(1),58–66 (2008).
    • 21  Stankus JJ, Soletti L, Fujimoto K, Hong Y, Vorp DA, Wagner WR: Fabrication of cell microintegrated blood vessel constructs through electrohydrodynamic atomization. Biomaterials28(17),2738–2746 (2007).
    • 22  Nieponice A, Soletti L, Guan J et al.: Development of a tissue-engineered vascular graft combining a biodegradable scaffold, muscle-derived stem cells and a rotational vacuum seeding technique. Biomaterials29(7),825–833 (2008).
    • 23  Konig G, Mcallister TN, Dusserre N et al.: Mechanical properties of completely autologous human tissue engineered blood vessels compared with human saphenous vein and mammary artery. Biomaterials30(8),1542–1550 (2009).
    • 24  Kim BS, Putnam AJ, Kulik TJ, Mooney DJ: Optimizing seeding and culture methods to engineer smooth muscle tissue on biodegradable polymer matrices. Biotechnol. Bioeng.57(1),46–54 (1998).
    • 25  Soletti L, Hong Y, Guan J et al.: A bilayered elastomeric scaffold for tissue engineering of small diameter vascular grafts. Acta Biomater.6(1),110–122 (2010).
    • 26  George J, Onodera J, Miyata T: Biodegradable honeycomb collagen scaffold for dermal tissue engineering. J. Biomed. Mater. Res. A87(4),1103–1111 (2008).
    • 27  Genovese JA, Spadaccio C, Langer J, Habe J, Jackson J, Patel AN: Electrostimulation induces cardiomyocyte predifferentiation of fibroblasts. Biochem. Biophys. Res. Commun.370(3),450–455 (2008).
    • 28  Onodera N, Tamaki T, Okada Y, Akatsuka A, Aoki D: Identification of tissue-specific vasculogenic cells originating from murine uterus. Histochem. Cell Biol.125(6),625–635 (2006).
    • 29  Jaques LB, Sue TK, Mcduffie NM, Wice SM: Heparin and sulfated mucopolysaccharides – a micro system for quantitative differentiation and determination. Can. J. Physiol. Pharmacol.55(5),1179–1189 (1977).
    • 30  Yang Z, Birkenhauer P, Julmy F et al.: Sustained release of heparin from polymeric particles for inhibition of human vascular smooth muscle cell proliferation. J. Control Release60(2–3),269–277 (1999).
    • 31  Kent KC, Oshima A, Whittemore AD: Optimal seeding conditions for human endothelial cells. Ann. Vasc. Surg.6(3),258–264 (1992).
    • 32  Pham QP, Sharma U, Mikos AG: Electrospinning of polymeric nanofibers for tissue engineering applications: a review. Tissue Eng.12(5),1197–1211 (2006).▪ Examines the crucial role of heparin in endothelial cell adhesion and homeostasis, and provides clues regarding its capability to improve cell attachment to the scaffold, providing trophic and differentiating signaling and acting as a depot role in the ECM for the growth factors, improving their release and enhancing their stability against thermal denaturation and enzymatic digestion.
    • 33  Yoon JJ, Chung HJ, Lee HJ, Park TG: Heparin-immobilized biodegradable scaffolds for local and sustained release of angiogenic growth factor. J. Biomed. Mater. Res. A79(4),934–942 (2006).
    • 34  Cai S, Liu Y, Zheng Shu X, Prestwich GD: Injectable glycosaminoglycan hydrogels for controlled release of human basic fibroblast growth factor. Biomaterials26(30),6054–6067 (2005).
    • 35  Chung HJ, Kim HK, Yoon JJ, Park TG: Heparin immobilized porous PLGA microspheres for angiogenic growth factor delivery. Pharm. Res.23(8),1835–1841 (2006).
    • 36  Casper CL, Yamaguchi N, Kiick KL, Rabolt JF: Functionalizing electrospun fibers with biologically relevant macromolecules. Biomacromolecules6(4),1998–2007 (2005).
    • 37  Serizawa N, Takei Y, Okubo H, Yamasina S, Enomoto N, Sato N: Effect of low-molecular-weight heparin on the commitment of bone marrow cells to liver sinusoidal endothelial cells in CCL(4)-induced liver injury. Hepatol. Res.34(4),207–213 (2006).
    • 38  Centola M, Rainer A, Spadaccio C, De Porcellinis S, Genovese J, Trombetta M: Combining electrospinning and fused deposition modeling for the fabrication of a hybrid vascular graft. Biofabrication (2010) (In Press).
    • 39  Sank A, Wei D, Reid J et al.: Human endothelial cells are defective in diabetic vascular disease. J. Surg. Res.57(6),647–653 (1994).▪ Describes different functionalization techniques for tissue engineering, underlining the advantage of direct incorporation of biomolecules in the polymer solution in order to overcome the drawbacks of other methods, such as adsorption or microencapsulation.
    • 40  Guan J, Stankus JJ, Wagner WR: Biodegradable elastomeric scaffolds with basic fibroblast growth factor release. J. Control Release120(1–2),70–78 (2007).
    • 41  Chabut D, Fischer AM, Colliec-Jouault S et al.: Low molecular weight fucoidan and heparin enhance the basic fibroblast growth factor-induced tube formation of endothelial cells through heparan sulfate-dependent a6 overexpression. Mol. Pharmacol.64(3),696–702 (2003).▪ Contains relevant data concerning the heparin dosage capable of inducing endothelial differentiation in culture conditions.
    • 42  Bezuidenhout D, Davies N, Black M, Schmidt C, Oosthuysen A, Zilla P: Covalent surface heparinization potentiates porous polyurethane scaffold vascularization. J. Biomater. Appl.24(5),401–418 (2008).▪ Reports the effect of heparin in the inhibition of smooth muscle proliferation and intimal hyperplasia, allowing speculation on the possible advantages of the proposed heparin functionalized poly-L-lactide scaffold in the control of the phenomena recognized to be at the root of graft restenosis.
    • 43  Glagov S: Intimal hyperplasia, vascular modeling, and the restenosis problem. Circulation89(6),2888–2891 (1994).
    • 44  Edelman ER, Nathan A, Katada M, Gates J, Karnovsky MJ: Perivascular graft heparin delivery using biodegradable polymer wraps. Biomaterials21(22),2279–2286 (2000).