Aging is coupled with a decline of immune function, loss of bone mass and accumulation of marrow fat. Peroxisome proliferator-activated receptor-gamma (PPAR?) is a key factor regulating fat and bone cell formation. Studies show that PPAR? insufficiency can increase bone mass, suggesting that targeting PPAR? could be a novel strategy for new anti-osteoporosis therapies. However, it is not clear how PPAR? insufficiency increases bone because deletion of PPAR? gene in fat tissue also increases bone mass, and that global reduction of fat causes severe diabetes and insulin resistance. We hypothesize that bone-specific PPAR? inactivation, which blocks only bone marrow fat formation, will eliminate marrow fat-generated inflammatory responses, including inflammatory cytokines and inflammatory cells, increases the availability and activity of osteoblasts, and improves immune function, thereby reducing the pace of bone loss in aging. This hypothesis is formulated on the basis that marrow fat increases with aging and fat tissue produces large quantities of inflammatory cytokines. In addition to inhibit bone cell development, these cytokines are predicted to degrading the bone marrow microenvironment (causing chronic inflammation) and inhibiting the hematopoiesis resulting in a shift of progenitor cell from a predominant lymphoid to a myeloid proportion (skewing of hematopoiesis) because bone marrow is the place where immune system is originated. We will test our hypothesis using two PPAR? knockout mouse models, one blocks only bone marrow fat cell formation and the other reduces whole body fat mass.
Two specific aims are proposed.
Aims 1 will test the specific hypothesis that PPAR? controls adult bone homeostasis but not skeletal development or growth, and Aim 2 will test the specific hypothesis that inactivation of PPAR? eliminates fat-generated inflammatory responses, including cytokines and inflammatory cells, and boosts the immune function, which declines with aging and may contribute to age-induced bone loss. Results of this research will shed new light on our current understanding of the mechanisms by which PPAR? regulates bone metabolism and help develop new anti-osteoporosis therapies.

Public Health Relevance

With advancing age, bone marrow fat increases (fatty marrow) and bone mass and immune function decrease. Fat tissue produces large amounts of inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-?) and interleukin (IL)-1 and -6. These cytokines not only play a key role in the regulation of bone-forming osteoblast (inhibition) and bone-resorbing osteoclast (stimulation), but also play a central role in the regulation of immune and inflammatory responses. Many aging related diseases, including osteoporosis, are tightly associated with chronic inflammation and are stated accurately by a new word 'inflammaging' (inflamm + aging). A transcription factor named PPAR? is a key controlling fat cell formation. Because both the bone cells and marrow fat cells derive from a common bone marrow stem cell, PPAR? also regulates bone formation. However, the underlying mechanism by which PPAR? controls bone is not clear. This study will investigate how PPAR? causes bone loss and decline of immune function through controlling the bone marrow fat. We will use sophisticated bone-specific PPAR? gene knockout mouse models and the state-of-the-art immunology and molecular biology technologies to achieve these goals. Results of this research will shed new light on our current understanding of aging-induced bone loss and help develop new anti-osteoporosis therapies.

Agency
National Institute of Health (NIH)
Institute
National Institute on Aging (NIA)
Type
Research Project (R01)
Project #
3R01AG046248-04S1
Application #
9231194
Study Section
Special Emphasis Panel (ZAG1)
Program Officer
Williams, John
Project Start
2013-09-30
Project End
2018-04-30
Budget Start
2016-05-15
Budget End
2017-04-30
Support Year
4
Fiscal Year
2016
Total Cost
Indirect Cost
Name
Georgia Regents University
Department
Neurosciences
Type
Schools of Medicine
DUNS #
City
Augusta
State
GA
Country
United States
Zip Code
30912
Su, Yun; Shen, Xiaona; Chen, Jie et al. (2018) Differentially expressed genes in PPAR?-deficient MSCs. Mol Cell Endocrinol 471:97-104
Yang, Nianlan; Baban, Babak; Isales, Carlos M et al. (2017) Role of glucocorticoid-induced leucine zipper (GILZ) in inflammatory bone loss. PLoS One 12:e0181133
Baban, Babak; Yin, Lin; Qin, Xu et al. (2017) The role of GILZ in modulation of adaptive immunity in a murine model of myocardial infarction. Exp Mol Pathol 102:408-414
Yang, Nianlan; Baban, Babak; Isales, Carlos M et al. (2015) Crosstalk between bone marrow-derived mesenchymal stem cells and regulatory T cells through a glucocorticoid-induced leucine zipper/developmental endothelial locus-1-dependent mechanism. FASEB J 29:3954-63
Cao, Jay; Ou, Guomin; Yang, Nianlan et al. (2015) Impact of targeted PPAR? disruption on bone remodeling. Mol Cell Endocrinol 410:27-34
Pan, Guodong; Cao, Jay; Yang, Nianlan et al. (2014) Role of glucocorticoid-induced leucine zipper (GILZ) in bone acquisition. J Biol Chem 289:19373-82