Non-healing skeletal defects are addressed in over 2.2 million surgical cases each year. Bone morphogenetic protein 2 (BMP2), the main growth factor for bone regeneration, has significant adverse effects at the FDA approved dose for human use. These effects include life-threatening cervical swelling and promotion of adipogenesis or cyst-like bone voids from dose-dependent peroxisome proliferator-activated receptor ? (PPAR ?) upregulation. Thus, a critical barrier to progress in improving the safety and efficacy of BMP2 is development of molecular signaling strategies that effectively target osteogenesis induction and adipogenesis suppression to optimize bone formation. The current New Investigator resubmission addresses this barrier using NELL-1 (Nel-like molecule, type I). NELL-1 is a secreted factor that induces significant in vivo bone formation in multiple small and large animal models. NELL-1 represses BMP2-induced adipogenesis and augments BMP2-induced osteogenesis. Moreover, our new data indicate that NELL-1 activates Wnt/?-catenin signaling and suppresses PPAR ? signaling. This has led to our central hypothesis that NELL-1 improves the efficacy of BMP2-induced bone formation through: [1] activation of Wnt/ ? -catenin signaling, and [2] suppression of PPAR ? signaling, tested in four specific aims.
In AIM 1, we will determine the involvement of Wnt/ ? -catenin and PPAR ? signaling in NELL-1+BMP2 mediated bone repair. Using our published rodent femoral segmental defect (FSD) model, we will precisely evaluate Wnt and PPAR ? signaling in TOPgal Wnt reporter mice. Next in AIM 2, we will evaluate the necessity of Wnt/ ? -catenin signaling in NELL-1+BMP2 regulated bone healing. Here, we will induce Wnt/ ? -catenin 'loss of function' by biochemical (Wnt inhibitor) or RNAi (? -catenin shRNA) methods in our FSD model.
In AIM 3, we will determine if increased Wnt/ ? -catenin signaling is sufficient to reproduce NELL-1's effects on promoting BMP2-induced osteogenesis and inhibiting BMP2-induced adipogenesis. Here, we will induce Wnt/ ? -catenin gain of function by genetic means using Axin2-/- (null mice) and examine its effects in our FSD model. Finally in AIM 4, we will determine if decreased PPAR ? signaling can reproduce NELL-1's effects on promoting BMP2-induced osteogenesis and inhibiting BMP2-induced adipogenesis. PPAR ? loss of function will be induced using PPAR ? +/- mice and RNAi (PPAR ? shRNA) methods in our FSD model. Successful completion of the AIMS will improve efficacy and reduce adverse effects for BMP2 based skeletal regeneration based on the combination therapeutic NELL-1+BMP2. Moreover, increased induction of osteogenesis and suppression of adipogenesis among mesenchymal stem cells will lead to marked improvements in clinical bone repair.

Public Health Relevance

Non-healing bone defects are addressed in over 2.2 million surgical cases worldwide each year. Bone Morphogenetic Protein2 (BMP2), the most commonly used bone growth factor, has significant adverse effects including formation of poor quality, fatty bone. Our innovative solution uses NELL-1, a novel bone forming molecule to inhibit fatty bone formation and to improve the efficacy of BMP2-based bone regeneration therapies.

Agency
National Institute of Health (NIH)
Institute
National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS)
Type
Research Project (R01)
Project #
5R01AR061399-04
Application #
8856500
Study Section
Musculoskeletal Tissue Engineering Study Section (MTE)
Program Officer
Wang, Fei
Project Start
2012-07-01
Project End
2017-05-31
Budget Start
2015-06-01
Budget End
2016-05-31
Support Year
4
Fiscal Year
2015
Total Cost
$346,500
Indirect Cost
$121,500
Name
University of California Los Angeles
Department
Orthopedics
Type
Schools of Medicine
DUNS #
092530369
City
Los Angeles
State
CA
Country
United States
Zip Code
90095
Zhang, Yulong; Zheng, Zhong; Yu, Mengliu et al. (2018) Using an Engineered Galvanic Redox System to Generate Positive Surface Potentials that Promote Osteogenic Functions. ACS Appl Mater Interfaces 10:15449-15460
Li, Chenshuang; Zheng, Zhong; Jiang, Jie et al. (2018) Neural EGFL-Like 1 Regulates Cartilage Maturation through Runt-Related Transcription Factor 3-Mediated Indian Hedgehog Signaling. Am J Pathol 188:392-403
Shen, Jia; Chen, Xuepeng; Jia, Haichao et al. (2018) Effects of WNT3A and WNT16 on the Osteogenic and Adipogenic Differentiation of Perivascular Stem/Stromal Cells. Tissue Eng Part A 24:68-80
Meyers, Carolyn A; Xu, Jiajia; Zhang, Lei et al. (2018) Early Immunomodulatory Effects of Implanted Human Perivascular Stromal Cells During Bone Formation. Tissue Eng Part A 24:448-457
Tanjaya, Justine; Lord, Elizabeth L; Wang, Chenchao et al. (2018) The Effects of Systemic Therapy of PEGylated NEL-Like Protein 1 (NELL-1) on Fracture Healing in Mice. Am J Pathol 188:715-727
Lee, Soonchul; Wang, Chenchao; Pan, Hsin Chuan et al. (2017) Combining Smoothened Agonist and NEL-Like Protein-1 Enhances Bone Healing. Plast Reconstr Surg 139:1385-1396
Shi, J; Lee, S; Pan, H C et al. (2017) Association of Condylar Bone Quality with TMJ Osteoarthritis. J Dent Res 96:888-894
Li, Chenshuang; Jiang, Jie; Zheng, Zhong et al. (2017) Neural EGFL-Like 1 Is a Downstream Regulator of Runt-Related Transcription Factor 2 in Chondrogenic Differentiation and Maturation. Am J Pathol 187:963-972
Guo, Mian; James, Aaron W; Kwak, Jin Hee et al. (2016) Cyclophilin A (CypA) Plays Dual Roles in Regulation of Bone Anabolism and Resorption. Sci Rep 6:22378
Lee, Soonchul; Shen, Jia; Pan, Hsin Chuan et al. (2016) Calvarial Defect Healing Induced by Small Molecule Smoothened Agonist. Tissue Eng Part A 22:1357-1366

Showing the most recent 10 out of 21 publications