PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

Similar documents
Award Number: W81XWH TITLE: Regenerative Stem Cell Therapy for Breast Cancer Bone Metastasis

CONTRACTING ORGANIZATION: University of California Lawrence Berkeley National Laboratory Berkeley, CA 94720

TITLE: Dietary Genistein and Prostate Cancer Chemoprevention

TITLE: Genes Associated with Food Allergy and Eosinophilic Esophagitis

TITLE: Role of ADAM15 in Tumor/Endothelial Interactions Prostate Cancer Regression

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: New Advanced Technology to Improve Prediction and Prevention of Type 1 Diabetes

TITLE: A Tissue Engineering Approach to Study the Progression of Breast Tumor Metastasis in Bone

TITLE: Enhancing the Anti-Tumor Activity of ErbB Blockers with Histone Deaccetylase (HDAC) Inhibition in Prostate Cancer Cell Lines

TITLE: Neural Protein Synuclein (SNCG) in Breast Cancer Progression

TITLE: Breast Tumor-Generated Type 1 Collagen Breakdown Fragments Act as Matrikines to Drive Osteolysis

TITLE: Targeted Eradication of Prostate Cancer Mediated by Engineered Mesenchymal Stem Cells

CONTRACTING ORGANIZATION: Children s Hospital Los Angeles Los Angeles, CA 90027

TITLE: MODULATION OF T CELL TOLERANCE IN A MURINE MODEL FOR IMMUNOTHERAPY OF PROSTATIC ADENOCARCINOMA

TITLE: A PSCA Promoter Based Avian Retroviral Transgene Model of Normal and Malignant Prostate

TITLE: Effect of COX-2 (PGE2) and IL-6 on Prostate Cancer Bone Mets

TITLE: Properties of Leukemia Stem Cells in a Novel Model of CML Progression to Lymphoid Blast Crisis

CONTRACTING ORGANIZATION: North Eastern Ohio Universities Rootstown OH 44202

TITLE: Oxidative Stress, DNA Repair and Prostate Cancer Risk. PRINCIPAL INVESTIGATOR: Hua Zhao, Ph.D.

TITLE: Development of Antigen Presenting Cells for adoptive immunotherapy in prostate cancer

TITLE: Adipose Estrogen and Increased Breast Cancer Risk in Obesity: Regulation by Leptin and Insulin

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: Investigating the Role of TBX2 in the Inhibition of Senescence in Prostate Cancer

TITLE: Evaluation of DNA Methylation as a Target for Intraductal Therapy for Ductal Carcinoma in Situ of the Breast

Award Number: W81XWH

TITLE: Targeting the Immune System s Natural Response to Cell Death to Improve Therapeutic Response in Breast Cancers

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

Fort Detrick, Maryland

TITLE: Crosstalk Between Cancer Cells and Bones Via the Hedgehog Pathway Determines Bone Metastasis of Breast Cancer

AD (Leave blank) Award Number: W81XWH TITLE: Targeting Autophagy for the Treatment of TSC and LAM. PRINCIPAL INVESTIGATOR: Elizabeth Henske

TITLE: Notch in Pathological Angiogenesis and Lymphangiogenesis

TITLE: Harnessing GPR17 Biology for Treating Demyelinating Disease

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: Prevention and Treatment of Neurofibromatosis Type 1-Associated Malignant Peripheral Nerve Sheath Tumors

CONTRACTING ORGANIZATION: Sloan-Kettering Institute for Cancer Research New York, NY 10021

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

CONTRACTING ORGANIZATION: West Virginia University Morgantown, West Virginia

Award Number: W81XWH TITLE: Dietary Fish Oil in Reducing Bone Metastasis of Breast Cancer

Award Number: W81XWH TITLE: Characterizing an EMT Signature in Breast Cancer. PRINCIPAL INVESTIGATOR: Melanie C.

CONTRACTING ORGANIZATION: Regents of the University of Michigan Ann Arbor, MI 48109

TITLE: Induction of Ephs/Ephrins-Mediated Tumor Cells-Endothelial Cells Repulsion as an Anti-Cancer Therapeutic Approach

TITLE: The Role of HOX Proteins in Androgen-Independent Prostate Cancer

Award Number: W81XWH TITLE: Global Positioning Systems (GPS) Technology to Study Vector-Pathogen-Host Interactions

CONTRACTING ORGANIZATION: Oregon Health & Science University Portland OR 97239

TITLE: Investigation of the Akt/PKB Kinase in the Development of Hormone-Independent Prostate Cancer

AWARD NUMBER: W81XWH TITLE: Androgen Deprivation Therapy and Cognitive Impairment. PRINCIPAL INVESTIGATOR: Robert N. Pechnick, Ph.D.

Approved for public release; distribution unlimited

CONTRACTING ORGANIZATION: Mount Sinai School of Medicine New York, New York

TITLE: Systemic Oncolytic Cytokine HSV Therapy of Prostate Cancer. CONTRACTING ORGANIZATION: Massachusetts General Hospital Boston, MA

TITLE: Overcoming Resistance to Inhibitors of the Akt Protein Kinase by Modulation of the Pim Kinase Pathway

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

CONTRACTING ORGANIZATION: Western Institute For Biomedical Research Salt Lake City, UT

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: The Role Of Alternative Splicing In Breast Cancer Progression

La Jolla, CA Approved for Public Release; Distribution Unlimited

TITLE: Development of Antigen Presenting Cells for adoptive immunotherapy in prostate cancer

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

AWARD NUMBER: W81XWH TITLE: Treatment of Pain and Autonomic Dysreflexia in Spinal Cord Injury with Deep Brain Stimulation

TITLE: Interchromosomal Associations that Alter NF1 Gene Expression Can Modify Clinical Manifestations of Neurofibromatosis 1. Palo Alto, CA 94304

TITLE: 64Cu-DOTA-trastuzumab PET imaging in women with HER2 overexpressing breast cancer

AD (Leave blank) TITLE: Proteomic analyses of nipple fluid for early detection of breast cancer

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

CONTRACTING ORGANIZATION: Johns Hopkins University School of Medicine Baltimore, MD 21205

TITLE: Hyaluronic acid is overexpressed in fibrotic lung tissue and promotes collagen expression

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: Inhibitors of Histone Deacetylases for Radiosensitization of Prostate Cancer

TITLE: Identification of New Serum Biomarkers for Early Breast Cancer Diagnosis and Prognosis Using Lipid Microarrays.

Roles of microrna-mediated Drug Resistance in Tumor Stem Cells of Small Cell Lung Carcinoma

CONTRACTING ORGANIZATION: Spaulding Rehabilitation Hospital, Boston, MA 02129

TITLE: Computerized Tailored Interventions for Behavioral Sequelae of Post-Traumatic Stress Disorder in Veterans

CONTRACTING ORGANIZATION: University of Texas M.D. Anderson Cancer Center Houston, TX 77030

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

CONTRACTING ORGANIZATION: Joan & Sanford I. Weill Medical College of Cornell University New York, NY 10065

Award Number: W81XWH TITLE: A Novel Membrane-Permeable, Breast-Targeting, Pro-Apoptotic Peptide for Treatment of Breast Cancer

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland Approved for public release; distribution unlimited

CONTRACTING ORGANIZATION: Johns Hopkins Bloomberg School of Public Health

CONTRACTING ORGANIZATION: Cold Spring Harbor Laboratory Cold Spring Harbor, NY 11724

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: Development of Antigen Presenting Cells for adoptive immunotherapy in prostate cancer

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: Harnessing GPR17 Biology for Treating Demyelinating Disease

TITLE: Microenvironments and Signaling Pathways Regulating Early Dissemination, Dormancy, and Metastasis

Award Number: W81XWH

Page 1 AWARD NUMBER: W81XWH TITLE: A Novel Pleiotropic Anti-Inflammatory Drug to Reduce ARDS Incidence. PRINCIPAL INVESTIGATOR: Gary Nieman

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: "Impact of Obesity on Tamoxifen Chemoprevention in a Model of Ductal Carcinoma in Situ"

CONTRACTING ORGANIZATION: Vanderbilt University Medical Center Nashville, TN

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

Detection of Prostate Cancer Progression by Serum DNA Integrity

TITLE: Prostate Expression Databases: Gene Expression Resources for Comparative Studies of Prostate Carcinogenesis

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

Promote Adjustment during Reintegration following Deployment

TITLE: Effect of Reminder Telephone Calls on Mammography Compliance in High Risk

AD (Leave blank) PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

Transcription:

Award Number: W81XWH-15-1-0314 TITLE: Osteoimmune Mechanisms of Segmental Bone Fracture Healing and Therapy PRINCIPAL INVESTIGATOR: Selvarangan Ponnazhagan, Ph.D. CONTRACTING ORGANIZATION: The University of Alabama at Birmingham Birmingham, AL 35294 REPORT DATE: September 2016 TYPE OF REPORT: Annual PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland 21702-5012 DISTRIBUTION STATEMENT: Approved for Public Release; Distribution Unlimited The views, opinions and/or findings contained in this report are those of the author(s) and should not be construed as an official Department of the Army position, policy or decision unless so designated by other documentation.

REPORT DOCUMENTATION PAGE Form Approved OMB No. 0704-0188 Public reporting burden for this collection of information is estimated to average 1 hour per response, including the time for reviewing instructions, searching existing data sources, gathering and maintaining the data needed, and completing and reviewing this collection of information. Send comments regarding this burden estimate or any other aspect of this collection of information, including suggestions for reducing this burden to Department of Defense, Washington Headquarters Services, Directorate for Information Operations and Reports (0704-0188), 1215 Jefferson Davis Highway, Suite 1204, Arlington, VA 22202-4302. Respondents should be aware that notwithstanding any other provision of law, no person shall be subject to any penalty for failing to comply with a collection of information if it does not display a currently valid OMB control number. PLEASE DO NOT RETURN YOUR FORM TO THE ABOVE ADDRESS. 1. REPORT DATE (DD-MM-YYYY) September 2016 2. REPORT TYPE Annual 4. TITLE AND SUBTITLE Osteoimmune Mechanisms of Segmental Bone Fracture Healing and Therapy 3. DATES COVERED (From - To) 18 Aug 2015-17 Aug 2016 5a. CONTRACT NUMBER 5b. GRANT NUMBER W81XWH-15-1-0314 5c. PROGRAM ELEMENT NUMBER 6. AUTHOR(S) 5d. PROJECT NUMBER Selvarangan Ponnazhagan, Ph.D 5e. TASK NUMBER pons@uab.edu 5f. WORK UNIT NUMBER 7. PERFORMING ORGANIZATION NAME(S) AND ADDRESS(ES) 8. PERFORMING ORGANIZATION REPORT The University of Alabama at Birmingham Department of Pathology 1825, University Blvd., SHEL 814 Birmingham, AL 35294-0007 9. SPONSORING / MONITORING AGENCY NAME(S) AND ADDRESS(ES) 10. SPONSOR/MONITOR S ACRONYM(S) U.S. Army Medical Research and Materiel Command 504 Scott Street Fort Detrick MD 21702-5012 11. SPONSOR/MONITOR S REPORT NUMBER(S) 12. DISTRIBUTION / AVAILABILITY STATEMENT 13. SUPPLEMENTARY NOTES Distribution Unlimited 14. ABSTRACT The proposal addresses a very important area of bone fracture healing that is a major health and financial burden in the United States. Towards developing an effective approach to cure segmental bone fractures, it is essential to understand additional mechanisms that play a crucial role in bone healing through participation of cells other than osteoblasts, osteoclasts and their respective progenitors. Bone fractures heal with overlapping phases of inflammation, cell proliferation, and bone remodeling. Osteogenesis and angiogenesis are known to work in concert to control many stages of this process, but when one is impaired, it leads to failure of fracture healing. During fracture repair, there is an abundant infiltration of immune cells at the fracture site that not only mediate the inflammatory responses, but also exert influence on neovasculature. The proposed studies will characterize the role of IMC in healing segmental bone fracture in tibia using an immunocompetent mouse model. Studies will encompass a detailed analysis of IMC function in bone healing through enhancement of cellular and molecular signals on endothelial cell invasion, migration and proliferation, aiding in proper ossification and healing. Overall, the studies will test the hypothesis that signals provided by IMC play a crucial role in interfacing sequential events of angiogenesis and osteogenesis that are vital for proper bone healing. 15. SUBJECT TERMS Bone fracture; non-union defect; immature myeloid cells; angiogenesis 16. SECURITY CLASSIFICATION OF: 17. LIMITATION OF ABSTRACT Unclassified Unclassified 18. NUMBER OF PAGES 19a. NAME OF RESPONSIBLE PERSON USAMRMC a. REPORT b. ABSTRACT c. THIS PAGE 19b. TELEPHONE NUMBER Unclassified Unclassified Unclassified area code) (include Standard Form 298 (Rev. 8-98) 13 Prescribed by ANSI Std. Z39.18

Table of Contents Introduction... 4 BODY. 5 Key Research Accomplishments.. 13 Reportable Outcomes.. 13 Conclusions... 13 References.....N/A Appendices.....NA

Title of the Grant: Osteoimmune Mechanisms of Segmental Bone Fracture Healing and Therapy Award number: W81XWH-15-1-0314 Principal Investigator: Selvarangan Ponnazhagan, Ph.D. Annual Report: 10/1/2015-09/30/2016 INTRODUCTION The incidence of segmental fractures is significantly higher in military personnel compared to civilians. Despite efforts involving allografts, surgery and fixators, intramedullary nailing and invasive plate fixing to heal segmental fractures, unfortunately, lingering effects remain when the fracture involves long segmental bone loss. This necessitates the importance of identifying key molecular signals involved during critical phases of bone fracture healing, and developing novel therapies based on such understanding of molecular and cellular interactions during fracture healing. Large segmental bone defects are still a very serious problem in clinical practice. Especially when a significant quantity of bone is lost in the periosteum, it severely reduces the source of progenitors for remodeling. Current treatment options, especially for long segmental defects still pose complications of morbidity at the donor sites, devitalization of the transplant due to resorption process, leading to reduced mechanical stability - all contributing to difficulties in clinical management. To overcome these limitations, current efforts are focused on: tissue engineering approaches aimed at developing osteoconductive scaffolds, better quality synthetic bone grafts, and use of autologous mesenchymal stem cell transplantation. Although these are promising, the major focus still remains in enhancing osteogenic effects in the fracture area. In order to better approach the problem, it is apparent that key cellular and molecular signals that determine fracture healing during initial and crucial phases of bone healing need to be elucidated. Following fracture, there is disruption of normal vasculature that results in the release of growth factors by the damaged matrix and surrounding cells, signaling an influx of immune cells and osteogenic progenitors to remodel the fracture site. The initial injury to the blood vessels is contained to the surrounding area and progresses into a hematoma. Immune cells that infiltrate the hematoma to fight infection also secrete various cytokines and growth factors that are responsible for helping initiate the fracture repair cascade. Growth factors such as vascular endothelial growth factor (VEGF), bone morphogenetic proteins (BMP), transforming growth factor (TGF)-β, and cytokines such as interleukin (IL)-1 and -6 have been implicated in the migration of mesenchymal stem cells (MSC), and aid in recruitment of inflammatory cells to the fracture site. Once fracture healing is initiated, osteogenesis and angiogenesis work in concert to control many stages of bone remodeling. Fractures that do not heal are not avascular, but nonunion are more likely to occur when blood vessel growth is delayed after fracture. Therefore, understanding how the initial inflammatory reaction of fracture healing is coupled to signaling the fracture repair cascade will uncover mechanisms involved in the fate of bone healing, and will provide new opportunities for therapeutic intervention. To this end, the proposal seeks to characterize the roles of a key immune cell population at various stages of fracture healing in a large segmental bone defect model, and therapeutically use it in healing of nonunion segmental fracture in mice. SPECIFIC AIMS: 1. To determine molecular mechanisms of IMC function in healing of segmental bone fractures. 2. To determine therapeutic effects of IMC adoptive transfer in an immunocompetent mouse tibial segmental bone fracture model. 4

BODY Rationale and Approach: Bone fractures heal with overlapping phases of inflammatory influx of immune cells, cell proliferation, endochondral bone formation, coupled remodeling (osteogenesis and angiogenesis). The inflammatory phase characterized by an influx of immune cells and formation of a hematoma. Therefore we want to understand how the initial inflammatory reaction and the recruitment of immune cells at the site of fracture are linked to the fracture repair process. We hypothesize that identification of immune cell phenotypes and accompanying chemokines in fractured microenvironment promises to yield crucial information for designing strategies to treat non-union bone fractures. For instance we envision that identification of particular immune cell subtype(s) and/or chemokine(s) at site of bone fracture could be used as a bait to facilitate homing of other angiogenic and/or immune modulators to facilitate bone repair process. To accomplish this goal, in the first part of our project, we focus our efforts on to conduct an unbiased screen to investigate changes in diverse immune cell subsets in femoral bone marrow isolates obtained at critical time-points following a segmental fracture. We have created segmental fractures recently and analyzed data. The following sections provide highlights of results obtained: Segmental defect leads to systemic increase in B220+ cells at Day 5 and 10 in C57BL6 mice: We found that segmental fracture leads to a sharp and systemic (effect observed in both contralateral and fracture bearing femurs) increase in B220+ cells in bone marrow isolates at Day 5 and Day 10 following segmental fractures in immune-competent C57BL6 females (8-10 weeks old). To further identify the type of upregulated B220+ cells we also analyzed Plasmacytoid Dendritic cells and B-cell subpopulations. We found a sharp systemic increase in pdc s cell subpopulations at Day 5 and 10 post facture, time points during bone healing processes such as intramembranous bone formation and endochondral ossification are occurring (Fig 1). Further studies will be conducted to establish relationship between the marked upregulation of B220+ immune cell subpopulation and bone repair. Fig 1: BM isolates from C57BL6 mice bearing segmental fracture were analyzed by flow cytometry. A, Quantification of B220 positive and pdc s immune cell subpopulation from control, contralateral and segmental fracture bearing femurs. B, C. Representative FAC s contour images denoting the gating structure and systemic increase in B cell and pdc subpopulations at Day 5 and 10 (see inset) 5

Segmental defect leads to a marked systemic increase in Endothelial Progenitor Cells (EPC s) initially, followed by a rapid decline at later time-points: The screen for immune cell subpopulations also revealed a marked and systemic increase in EPC immune cell subpopulation following segmental fracture (Fig 2). This finding is particularly encouraging in the light of the observation that bone repair process requires angiogenic mediators. The systemic increase in EPCs as early as Day 1 post fracture may indicates that the BM is primed for angiogenic potential at a very early stage of the bone repair process. Furthermore we found that segmental fracture lead to a modest but a persistent increase in EPC subpopulations in SF femurs Fig 2: BM isolates from C57BL6 mice bearing segmental fracture were analyzed by Flow Cytometry. A, Quantification of EPC s from control, contralateral and segmental fracture bearing femurs. B, C. Representative FAC s contour images denoting the gating structure and systemic increase in EPC subpopulations at Day 5 and 10 (see inset) Dendritic cells show a systemic increase on Day 1 post-sf and rapid systemic decline at later time-points: Following critical size segmental defect we observed a sharp increase in Classical Dendritic Cells at very earl time points. Interestingly this increase is not sustained at later timepoints and we observed a systemic decline in Classical DC s (Fig 3). DC s are classical antigen presenting cells that interact with T-cells and B-cells and orchestrate the intensity of adaptive immune response. To assess if the effects in Classical DC s are mirrored in T-cells we next analyzed specific T-cell subpopulations 6

Fig 3: BM isolates from C57BL6 mice bearing segmental fracture were analyzed by Flow Cytometry. A, Quantification of classical DC s from control, contralateral and segmental fracture bearing femurs. B, C. Representative FAC s contour images denoting the gating structure and systemic increase in EPC subpopulations at Day 5 and 10 (see inset) Summary of changes in immune cell subtypes following segmental defects: We analyzed the data obtained in the last 6 months on endothelial cell and immune cell profiling at the fracture site following segmental bone fracture. The alterations observed in specific immune cell subtypes in in BM isolates of mice femurs bearing segmental defects are summarized in Table 1. The alterations are classified as persistent (observed at all-time points), local vs. systemic (occurring only in segmental defect femurs vs. changes occurring both in contralateral and segmental defects bearing femurs). Future studies will entail using BMD analysis, immunohistochemistry studies, cytokine profiling to correlate the bone healing process timeline with changes associated in immune cell subtypes. Immune cell subtype Effect observed Dendritic, Endothelial Progenitor B220+, plasmacytoid DCs Macrophages Neutrophils Sharp decline after increase on Day 1 Sharp increase on Day 10 Time-dependent decrease Time-dependent gradual increase Table 1: Changes in immune cell subtypes following segmental defects in C57BL6 female mice 7

Since bone fracture therapy will also require stimulation with bone morphogenetic protein (BMP)-2, our next focus has been to improve stability of this protein and to reduce proteasomal degradation. Towards this major goal, we performed studies to identify domains that are amenable for degradation by proteasomal enzymes. Following are data obtained in this regard. Proteasomal Blockade Stabilizes Intracellular BMP-2: To assess if proteasomal block causes an increase in BMP-2 precursor, we transfected 293T cells with a construct expressing human BMP-2 precursor under a strong CMV promoter. The cells were treated with two different proteasomal inhibitors post transfection. Intracellular BMP-2 shows dose dependent and time dependent stabilization following proteasomal block, indicating that abrogating conventional routes of intracellular protein degradation can enhance the BMP precursor protein level. Fig 4: A, B 293T cells were transfected with N-terminal HA-tagged pre-pro-bmp2 precursor with increasing doses of MG-132. Protein lysates from harvested cells were immunoblotted with HA antibody. C, D 293T cells were transfected with N-terminal HA-tagged pre-pro-bmp2 precursor with Epoximicin for indicated times. Protein lysates from harvested cells were immunoblotted with HA antibody. Pre-pro-BMP-2 exhibits a time-dependent increase in protein levels following proteasomal blockade Assessment of steady state turnover of BMP-2 precursor: We next assessed the steady state turnover of BMP-2 precursor protein. The cells were treated with Actinomycin D and Cycloheximide to block de-novo mrna and protein synthesis respectively. Following this treatment, short-term degradation kinetics of BMP-2 precursor protein was assessed in presence or absence of proteasomal blockade. We found proteasomal blockade substantially delayed BMP- 2 degradation, with dramatic improvement in its intracellular half-life as shown in figures below: 8

Fig 5: A,B 293T cells were transfected with N-terminal HA-tagged pre-pro-bmp2 precursor and degradation kinetic was followed after blocking de-novo protein synthesis (CHX). B, Densitometric quantification of immunoblot shown in Fig 5A. Epoximicin and to lesser extent MG-132 substantially delays BMP-2 degradation. Fig 6: A,B 293T cells were transfected with N-terminal HA-tagged pre-pro-bmp2 precursor and degradation kinetic was followed after blocking de-novo mrna synthesis (CHX) B Densitometric quantification of immunoblot shown in Fig 6A. 9

Immature myeloid cells increase endothelial cell migration and motility: It has previously been shown that a subpopulation of differentiated CD34 - /CD14 + monocytes isolated from human peripheral blood will co-express endothelial-specific surface antigens and grow tube-like structures when cultured under angiogenic stimulation. Furthermore, when co-cultured with carcinoma cells, CD11b + Gr1 + cells have been shown to differentiate into endothelial cells, and incorporate into the tumor vasculature. Considering that angiogenesis and osteogenesis are strongly coupled during bone formation, and that characterization of normal IMC revealed a predominantly monocytic phenotype (Ly6C + Ly6G - ), we sought to determine if IMC in the bone fracture microenvironment displayed an angiogenic phenotype. To this end, IMC infiltrates isolated from the site of broken femur were cocultured with HUVEC in transwell migration, and motility assays in serum-free media to demonstrate factors from IMC were responsible for any changes in HUVEC phenotype. Results of this study are shown below: Cigure 7: HUVEC HUVEC+day-0 IMC HUVEC+day-3 IMC HUVEC+day-7 IMC IMC isolated from naïve mice or mice after segmental fractures on days 1, 3, and 7were co-cultured with HUVEC in serum-free medium. IMC were placed in the bottom chamber and HUVEC in the top of the Boyden chamber. HUVEC cells capable of passing though pores were fixed and stained with neutral red after 4 hours. Figure 7 above demonstrated that IMC, isolated on day-3 and day-7 following bone fracture have the potential to initiate migration of endothelial 40 cells. Quantitative analysis of the motility Figure 8: ** * 35 assay is given in Figure 8. There was a significant difference in the number of 30 HUVE cells that migrated through the 25 Boyden chamber when co-cultured with 20 IMC isolated on day-3 or day-7 following bone fracture. 15 Number of migrated cells/398 µm 2 10 Next, a scratch assay was performed. Briefly, HUVEC cells were grown to 5 confluence in 24-well plates and scratched 0 with the narrow end of a sterile p200 pipette Control Day 0 Day 3 Day 7 tip. Media was changed to remove floating cells and replaced with serum-free endothelial cell basal medium (Lonza). Photomicrographs were taken at 200x magnification immediately after initial wounding, and the scratch area was measured. Transwell inserts were inserted to the wells, and IMC were placed in the upper chamber in the same serum-free endothelial basal media. Cells were then incubated at 37 C with 5% CO 2. After 12 hours, photomicrographs were again taken at 200x magnification and the scratch area was analyzed using ImageJ (NIH). Results are shown in Figures 9 and 10. 10

Figure 9: HUVEC HUVEC + wt IMC HUVEC + day-3 IMC HUVEC + day-7 IMC 12 hrs 0 hr Figure 10: Percent migration 120 100 80 60 40 20 * * 0 hrs 12 hrs 0 HUVEC HUVEC + wt IMC HUVEC + day-3 IMC HUVEC + day-7 IMC These data further confirmed that activated IMC caused an increase in cell migration with significant migration observed in HUVEC, co-cultured with IMC isolated on days 3 and 7 after fracture. These results indicate IMC in the post-fracture milieu aid in endothelial cell recruitment during fracture repair. 11

Immature myeloid cells increase endothelial tube formation in vitro. To further elucidate the impact of IMC on angiogenesis in the bone fracture environment, we investigated whether isolated IMC could affect endothelial tube formation. IMC were isolated from wild-type and gemcitabine treated mice, both before and after segmental defect, and co-cultured with HUVEC on reduced growth factor (RGF) Matrigel. Naïve (control) and activated IMC attached to HUVEC cells and increased the number of branch points and the number of junctions significantly, compared to HUVEC cultured alone (Figures 11-13). This data confirmed IMC provide important paracrine effects on angiogenesis at the fracture site through soluble factors. Briefly, IMC isolated from naïve or 1, 3, or 7 days after segmental defect are cocultured directly with HUVEC. IMC bind directly to HUVEC. IMC were labeled with CFSE, and HUVEC were labeled with PKH26, 20x magnification (Figure 11).Representative images of HUVEC and IMC co-culture on tube formation assay are given in Figure 12. Quantitative analysis of tube formation using Angiogenesis Analyzer in ImageJ, n = 3 and three 10x images per well is shown in Figure 13 (*P<0.05). Figure 11: HUVEC Day-3 IMC Merge Figure 12: HUVEC HUVEC + day-0 IMC HUVEC + day-3 IMC HUVEC + day-7 IMC Figure 13: Number of branches/1540 µ 2 120 100 80 60 40 20 0 HUVEC HUVEC + ctrl IMC HUVEC + day-3 IMC * HUVEC + day-7 IMC 12

KEY RESEARCH ACCOMPLISHMENTS: Characterization of immune cells following bone fracture was performed by flow cytometry. Novel approaches are being undertaken to improve the stability of bone morphogenetic protein (BMP)-2 for therapeutic use in fracture healing. Functional assays were performed to characterize immature myeloid cells (IMC) from bone fracture site. The studies were performed to identify properties of IMC in endothelial cell mobilization and vascular remodeling. This is critical since the early phase of fracture healing is greatly influenced by angiogenic cascade. REPORTABLE OUTCOMES None CONCLUSIONS Immature myeloid cells appear to play a crucial role in promoting angiogenic signaling, which is important for initiating osteogenic response for fracture healing. PERSONNEL RECEIVING PAY FROM THIS GRANT Selvarangan Ponnazhagan, Ph.D. Joo Hyoung Lee, Ph.D. 13