Modulation of RUNX2 Activity by ER? in Osteoblasts The bone-sparing properties of estrogens are mediated by ER? in a variety of cell types, including cells of the osteoblast and osteoclast lineages, but the underlying molecular mechanisms are poorly understood. In particular, there are contrasting reports on how estradiol (E2) affects RUNX2, an osteoblast master regulator that has been implicated in human bone mass control through GWAS. RUNX2 plays critical roles in the osteoblast lineage to stimulate both autonomous cellular differentiation (and thus bone formation) and osteoblast-driven osteoclastogenesis (and thus bone resorption). While this project initiated with a focus on inhibition of RUNX2 by E2, the present renewal application aims to take genome-wide approaches to understand why E2 does not inhibit RUNX2-driven transcription uniformly. We show that RUNX2 activity upon various targets is modulated differently, with most targets inhibited, but others not inhibited and some even cooperatively stimulated, to various extents, by RUNX2 and E2. Locus-dependent differential modulation of RUNX2 is expected to ultimately change the balance between RUNX2-mediated osteoblast differentiation and RUNX2-mediated osteoblast-driven osteoclastogenesis. Our preliminary data also demonstrate that both raloxifen and lasofoxifene poorly mimic E2 in modulating activity of RUNX2 at different loci. Mechanisms underlying the locus- and ligand-dependent modulation of RUNX2 by ER? are completely unknown. Based on our preliminary results, we hypothesize that ER? differentially modulates RUNX2 across the osteoblast genome depending on local relative positions of sites occupied by RUNX2 and ER? and the ER ligand. Additionally, we hypothesize that local collaborating transcription factors (TFs), as well as specific cognate motif sequences for RUNX2, ER? and collaborating TFs shape local effects of ER? on RUNX2. We will first investigate by ChIP-seq analysis of histone marks how E2 modulates RUNX2-driven changes to the chromatin activation status at every genomic locus. The mutual effects of ER? and RUNX2 on occupying their target loci will also be determined genome-wide by ChIP-seq. Computational models will then be developed to explain the locus-specific combinatorial transcriptional regulation by RUNX2 and ER?. These models will be tested, first computationally and then experimentally, for their ability to predict effects of E2 on RUNX2 activity. Finally, since ER? in pre-osteoblasts protects female cortical bone in mice, and since raloxifen and lasofoxifene, SERMs commonly prescribed in the US and in Europe, respectively, only reduce the risk for vertebral (predominantly trabecular) osteoporotic fractures, but not non-vertebral fractures, we will also decode how raloxifene- and lasofoxifene-bound ER? modulate RUNX2 activity genome wide. Principal genomic determinants will be identified, which explain the differential modulation of RUNX2 activity as a function of the locus (different targets) and the ligand (E2 vs. raloxifen vs. lasofoxifene). This project will help understand how E2 differentially modulates RUNX2 activity at different loci to potentially alter the balance between bone formation and bone resorption. Gene sets will be identified where SERMs mimic, or do not mimic E2. While unveiling the underlying genomic codes, tools will be generated for future investigation of how E2, or any SERM, modulate RUNX2 activity at prototypic loci to differentially regulate specific gene sets.

Public Health Relevance

(Public Health Relevance Statement) Modulation of RUNX2 Activity by ER? in Osteoblasts Molecular mechanisms of estrogens in bone are poorly understood, and efficacy of estrogen-like drugs to prevent postmenopausal osteoporosis is limited. This project will test a rich network of interactions in bone cells between estrogen signaling and RUNX2, a master regulator of bone formation, bone resorption and bone mass control. A comparison with currently available estrogen-like drugs will be carried out to explain their limitations.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
High Priority, Short Term Project Award (R56)
Project #
2R56DK071122-08A1
Application #
9503274
Study Section
Skeletal Biology Structure and Regeneration Study Section (SBSR)
Program Officer
Malozowski, Saul N
Project Start
2005-04-01
Project End
2019-07-31
Budget Start
2017-08-22
Budget End
2019-07-31
Support Year
8
Fiscal Year
2017
Total Cost
Indirect Cost
Name
University of Southern California
Department
Orthopedics
Type
Schools of Medicine
DUNS #
072933393
City
Los Angeles
State
CA
Country
United States
Zip Code
90033
Amzaleg, Yonatan; Ji, Jie; Kittivanichkul, Donlaporn et al. (2018) Estrogens and selective estrogen receptor modulators differentially antagonize Runx2 in ST2 mesenchymal progenitor cells. J Steroid Biochem Mol Biol 183:10-17
Costello, Kevin R; Schones, Dustin E (2018) Chromatin modifications in metabolic disease: Potential mediators of long-term disease risk. Wiley Interdiscip Rev Syst Biol Med 10:e1416
Luo, Jiao; Chimge, Nyam-Osor; Zhou, Beiyun et al. (2018) CLDN18.1 attenuates malignancy and related signaling pathways of lung adenocarcinoma in vivo and in vitro. Int J Cancer 143:3169-3180
Martin, Anthony; Yu, Jiali; Xiong, Jian et al. (2017) Estrogens and androgens inhibit association of RANKL with the pre-osteoblast membrane through post-translational mechanisms. J Cell Physiol 232:3798-3807
Morimoto, Eri; Li, Meng; Khalid, Aysha B et al. (2017) Glucocorticoids Hijack Runx2 to Stimulate Wif1 for Suppression of Osteoblast Growth and Differentiation. J Cell Physiol 232:145-53
White, James; Jago, Russell; Thompson, Janice L (2014) Dietary risk factors for the development of insulin resistance in adolescent girls: a 3-year prospective study. Public Health Nutr 17:361-8
Baniwal, Sanjeev K; Chimge, Nyam-Osor; Jordan, V Craig et al. (2014) Prolactin-induced protein (PIP) regulates proliferation of luminal A type breast cancer cells in an estrogen-independent manner. PLoS One 8:e62361
Koromila, Theodora; Baniwal, Sanjeev K; Song, Yae S et al. (2014) Glucocorticoids antagonize RUNX2 during osteoblast differentiation in cultures of ST2 pluripotent mesenchymal cells. J Cell Biochem 115:27-33
White, James; Shelton, Katherine H; Elgar, Frank J (2014) Prospective associations between the family environment, family cohesion, and psychiatric symptoms among adolescent girls. Child Psychiatry Hum Dev 45:544-54
Börjesson, Anna E; Farman, Helen H; Engdahl, Cecilia et al. (2013) The role of activation functions 1 and 2 of estrogen receptor-? for the effects of estradiol and selective estrogen receptor modulators in male mice. J Bone Miner Res 28:1117-26

Showing the most recent 10 out of 25 publications