Return to search

In vitro and in vivo growth factor delivery to chondrocytes and bone-marrow-derived stromal cells in cartilage and in self-assembling peptide scaffolds

Thesis (Ph. D.)--Massachusetts Institute of Technology, Dept. of Biological Engineering, 2010. / This electronic version was submitted by the student author. The certified thesis is available in the Institute Archives and Special Collections. / Vita. Cataloged from student-submitted PDF version of thesis. / Includes bibliographical references. / The inability of articular cartilage to repair itself after acute injury has been implicated in the development of osteoarthritis. The objective of this work was to develop methods for delivering growth factors to cartilage and to test the ability of a self-assembling peptide scaffold, (KLDL)3, with or without growth factors to augment repair. Delivery methods included growth factor adsorption, scaffold-tethering, and modification of growth factor structure. (KLDL)3 was modified to deliver IGF-1 and TGF-[beta]1 to chondrocytes and bone marrow- derived stromal cells (BMSCs), respectively, by adsorption and by biotin-streptavidin tethering. This study showed that while TGF-[beta]1 can be effectively delivered by adsorption, IGF-1 can not. Additionally, while tethering these factors provided longterm sequestration, tethering did not stimulate proteoglycan production in vitro. A full-thickness, critically sized, rabbit cartilage defect model was used to test the ability of (KLDL)3 with or without chondrogenic factors (TGF-[beta]1, dexamethasone, and IGF-1) and BMSCs to stimulate cartilage regeneration in vivo. (KLDL)3 alone showed the greatest repair after 12 weeks with significantly higher Safranin-O, collagen II immunostaining, and cumulative histology scores compared to untreated contralateral controls. Ongoing studies include the evaluation of (KLDL)3 in a clinically relevant sized equine defect co-treated with micro-fracture and subjected to strenuous exercise. A fusion protein was created by adding a heparin-binding domain to IGF-1 (HBIGF- 1), converting IGF-1 from a short-acting growth factor to one that can be retained and locally delivered in articular cartilage in vivo. It was shown that HB-IGF-1 is retained in cartilage through binding to negatively charged glycosaminoglycan chains, with chondroitin sulfate the most prevalent type in cartilage. HB-IGF-1 was shown to bind adult human cartilage and to be preferentially delivered and retained in rat articular cartilage after intra-articular injection. In contrast, unmodified IGF-1 was not detectable after intra-articular injection. These results suggest that modification of growth factors with heparin-binding domains may be a clinically relevant strategy for local delivery to cartilage. Taken together, these results show that (KLDL)3 self-assembling peptide hydrogels are customizable for growth factor delivery and can promote cartilage repair in vivo. In addition, the fusion protein HB-IGF-1 is preferentially retained in cartilage tissue compared to un-modified IGF-1. / by Rachel E. Miller. / Ph.D.

Identiferoai:union.ndltd.org:MIT/oai:dspace.mit.edu:1721.1/60997
Date January 2010
CreatorsMiller, Rachel E. (Rachel Elizabeth)
ContributorsAlan J. Grodzinsky., Massachusetts Institute of Technology. Dept. of Biological Engineering., Massachusetts Institute of Technology. Dept. of Biological Engineering.
PublisherMassachusetts Institute of Technology
Source SetsM.I.T. Theses and Dissertation
LanguageEnglish
Detected LanguageEnglish
TypeThesis
Format144 p., application/pdf
RightsMIT theses are protected by copyright. They may be viewed, downloaded, or printed from this source but further reproduction or distribution in any format is prohibited without written permission., http://dspace.mit.edu/handle/1721.1/7582

Page generated in 0.0106 seconds