Osteoporosis is a bone disease affecting nearly 1 in 3 women worldwide and over 55% of all Americans resulting in health care costs exceeding 19 billion dollars annually in the United States alone. Since the discovery of the TGF? inducible early gene-1 (TIEG) by our laboratory, we have demonstrated that loss of TIEG expression in mice results in multiple defects in osteoblasts and osteoclasts and confers a gender specific osteopenic phenotype affecting only female animals. Recently, TIEG has been identified as one of only a few genes whose altered expression levels or allelelic variations associate with decreased bone mass and osteoporosis in humans. During the past funding period, we have shown that TIEG expression is essential for estrogen signaling in bone and have identified a novel role for TIEG in mediating canonical Wnt signaling in the skeleton. These observations are of significant interest since estrogen remains one of the most important regulators of bone homeostasis in both men and women and since Wnt signaling is essential for normal bone development and maintenance. For these reasons, it is crucial to understand the regulation of these pathways in the skeleton. In this proposal, we present evidence that TIEG enhances the Wnt signaling pathway via dual mechanisms whereby it directly suppresses the expression of sclerostin and serves as a transcriptional co- activator for ?-catenin. Furthermore, our data suggest that TIEG serves as a direct link between the estrogen and canonical Wnt pathways in bone. Based on these observations, our central hypothesis is that TIEG is a crucial component of canonical Wnt signaling and serves a central role in regulating cross-talk between the estrogen and Wnt pathways in the skeleton. In order to test this hypothesis, the following Specific Aims are proposed: 1): Characterize the regulation of sclerostin gene expression by TIEG and determine its contribution to the observed osteopenic phenotype of TIEG knockout mice;2): Characterize the ability of TIEG to enhance canonical Wnt signaling through co-activation of ?-catenin;and 3) Determine if the gender specific (female only) osteopenic phenotype of TIEG KO mice results from defective cross-talk between the estrogen and canonical Wnt signaling pathways in bone. To address these Specific Aims, we will employ in vitro approaches to identify the molecular mechanisms by which TIEG suppresses sclerostin expression in osteocytes and enhances ?-catenin function in osteoblasts. Additionally, we will utilize multiple in vivo models and approaches to dissect the role of TIEG in regulating canonical Wnt pathway activity and cross-talk between estrogen and Wnt signaling in the skeleton. Considering the fundamental roles of estrogen and Wnt signaling in bone, as well as the present day targeting of these pathways for the treatment of osteoporosis, it is essential to understand the role of TIEG in regulating these two pathways. Completion of the proposed studies is therefore expected to provide significant knowledge with regard to developing novel strategies to treat this debilitating disease.

Public Health Relevance

Osteoporosis is a skeletal disease characterized by decreased bone mineral density and altered bone microarchitecture which ultimately results in bone fractures. Osteoporosis affects approximately 55% of all Americans resulting in health care costs of nearly 19 billion dollars annually, a figure that is expected to surpass 25 billion dollars by the year 2025. This project involves determining the role of TIEG, one of only a handful of genes identified to be clinically associated with the development of osteoporosis in humans, in regulating the canonical Wnt and estrogen signaling pathways in bone. Outcomes of the proposed studies are expected to further our understanding of osteoporosis and aid in the development of novel therapies to treat this debilitating disease.

Agency
National Institute of Health (NIH)
Institute
National Institute of Dental & Craniofacial Research (NIDCR)
Type
Research Project (R01)
Project #
2R01DE014036-10
Application #
8369584
Study Section
Skeletal Biology Structure and Regeneration Study Section (SBSR)
Program Officer
Wan, Jason
Project Start
2001-09-01
Project End
2017-06-30
Budget Start
2012-07-01
Budget End
2013-06-30
Support Year
10
Fiscal Year
2012
Total Cost
$408,350
Indirect Cost
$144,550
Name
Mayo Clinic, Rochester
Department
Type
DUNS #
006471700
City
Rochester
State
MN
Country
United States
Zip Code
55905
Subramaniam, Malayannan; Pitel, Kevin S; Bruinsma, Elizabeth S et al. (2018) TIEG and estrogen modulate SOST expression in the murine skeleton. J Cell Physiol 233:3540-3551
Marrero-Rodríguez, Daniel; Taniguchi-Ponciano, Keiko; Subramaniam, Malayannan et al. (2018) Krüppel-Like Factor 10 participates in cervical cancer immunoediting through transcriptional regulation of Pregnancy-Specific Beta-1 Glycoproteins. Sci Rep 8:9445
Farr, Joshua N; Weivoda, Megan M; Nicks, Kristy M et al. (2018) Osteoprotection Through the Deletion of the Transcription Factor Ror? in Mice. J Bone Miner Res 33:720-731
Subramaniam, Malayannan; Cicek, Muzaffer; Pitel, Kevin S et al. (2017) TIEG1 modulates ?-catenin sub-cellular localization and enhances Wnt signaling in bone. Nucleic Acids Res 45:5170-5182
Mishra, Vivek Kumar; Subramaniam, Malayannan; Kari, Vijayalakshmi et al. (2017) Krüppel-like Transcription Factor KLF10 Suppresses TGF?-Induced Epithelial-to-Mesenchymal Transition via a Negative Feedback Mechanism. Cancer Res 77:2387-2400
Najafova, Zeynab; Tirado-Magallanes, Roberto; Subramaniam, Malayannan et al. (2017) BRD4 localization to lineage-specific enhancers is associated with a distinct transcription factor repertoire. Nucleic Acids Res 45:127-141
Kammoun, Malek; Meme, Sandra; Meme, William et al. (2017) Impact of TIEG1 on the structural properties of fast- and slow-twitch skeletal muscle. Muscle Nerve 55:410-416
Weng, C-C; Hawse, J R; Subramaniam, M et al. (2017) KLF10 loss in the pancreas provokes activation of SDF-1 and induces distant metastases of pancreatic ductal adenocarcinoma in the KrasG12D p53flox/flox model. Oncogene 36:5532-5543
Kammoun, Malek; Pouletaut, Philippe; Canon, Francis et al. (2016) Impact of TIEG1 Deletion on the Passive Mechanical Properties of Fast and Slow Twitch Skeletal Muscles in Female Mice. PLoS One 11:e0164566
Delaney, Abigail A; Khan, Zaraq; Zheng, Ye et al. (2016) KLF10 Mediated Epigenetic Dysregulation of Epithelial CD40/CD154 Promotes Endometriosis. Biol Reprod 95:62

Showing the most recent 10 out of 38 publications