Estrogen loss increases osteoclastogenesis, decreases osteoclast and increases osteoblast apoptosis, and? causes loss of bone by decreasing anti-oxidant defense in these cell types. In addition, an age-dependent? decrease in bone strength and mass in ovary-intact C57BL/6 mice is temporally associated with an agedependent? increase in the prevalence of osteoblast apoptosis, decreased glutathione reductase (GSR)? activity and corresponding increases in the production of reactive oxygen species (ROS) in the bone marrow? and the phosphorylation of p66shc in vertebrae. Based on this evidence, it is hypothesized that increased? ROS levels with advancing age is a fundamental mechanism of the age-dependent decline of bone strength? and mass, and loss of estrogens exaggerates the adverse effects of aging on bone by decreasing defense? against ROS, thereby, contributing perpetually to the loss of bone mass and strength that persists for? decades after menopause, and is associated with old age. Estrogens enhance defense against ROS by? modulating the activity of anti-oxidant enzymes, attenuating ROS-mediated induction of cytokines, and? modulating signaling cascades that have been implicated in the defense against oxidative stress, apoptosis,? and aging. All these effects are mediated by extranuclear (nongenotropic) actions of the ER on cytoplasmic? kinases and result in changes in the birth and lifespan of osteoblasts, osteoclasts, and osteocytes. To test? these interrelated hypotheses, in vitro studies are proposed to investigate whether the effects of estrogens? on osteoblast and osteoclast apoptosis, as well as on osteoclastogenesis, are, at least in part, a direct? consequence of the ability of these hormones to: a) antagonize the actions of ROS by increasing the levels? of glutathione; and/or b) counteract ROS-activated signals on cytoplasmic kinases, and/or downstream? transcription factors and the resulting upregulation of cytokines. Further, the role of oxidative stress in the? molecular and cellular mechanism responsible for the loss of bone strength and mass associated with aging? and the extent to which estrogen deficiency and the resulting attenuation of defense against oxidative stress? in osteoblasts or osteoclasts contributes to these changes will be studied in mice over-expressing GSR in? osteoblasts or osteoclasts. Finally, mice in which ERa will be deleted from osteoblasts or osteoclasts by? means of CreXLoxP recombination; mice heterozygotes for an ERa knock-in mutant which cannot bind to? ERE; and mice over-expressing an ERa mutant, which is incapable of membrane localization and prevents? kinase-mediated signaling of the endogenous receptor, will be used to examine whether the antioxidant? actions of estrogens on osteoblasts and osteoclasts in vivo are ER-dependent and nongenotropic. This work? should help us understand how women become increasingly more susceptible to fractures as they grow old.? Moreover, it mav lead to the discovery of more effective treatments or even a cure for osteoporosis.

Agency
National Institute of Health (NIH)
Institute
National Institute on Aging (NIA)
Type
Research Program Projects (P01)
Project #
2P01AG013918-11
Application #
7094994
Study Section
Special Emphasis Panel (ZAG1-ZIJ-5 (J2))
Project Start
2006-06-01
Project End
2011-05-31
Budget Start
2006-06-01
Budget End
2007-05-31
Support Year
11
Fiscal Year
2006
Total Cost
$214,937
Indirect Cost
Name
University of Arkansas for Medical Sciences
Department
Type
DUNS #
122452563
City
Little Rock
State
AR
Country
United States
Zip Code
72205
Farr, Joshua N; Almeida, Maria (2018) The Spectrum of Fundamental Basic Science Discoveries Contributing to Organismal Aging. J Bone Miner Res 33:1568-1584
Iyer, Srividhya; Han, Li; Ambrogini, Elena et al. (2017) Deletion of FoxO1, 3, and 4 in Osteoblast Progenitors Attenuates the Loss of Cancellous Bone Mass in a Mouse Model of Type 1 Diabetes. J Bone Miner Res 32:60-69
Almeida, Maria; Laurent, Michaƫl R; Dubois, Vanessa et al. (2017) Estrogens and Androgens in Skeletal Physiology and Pathophysiology. Physiol Rev 97:135-187
Piemontese, Marilina; Almeida, Maria; Robling, Alexander G et al. (2017) Old age causes de novo intracortical bone remodeling and porosity in mice. JCI Insight 2:
Weinstein, Robert S; Hogan, Erin A; Borrelli, Michael J et al. (2017) The Pathophysiological Sequence of Glucocorticoid-Induced Osteonecrosis of the Femoral Head in Male Mice. Endocrinology 158:3817-3831
Kim, Ha-Neui; Chang, Jianhui; Shao, Lijian et al. (2017) DNA damage and senescence in osteoprogenitors expressing Osx1 may cause their decrease with age. Aging Cell 16:693-703
Ucer, Serra; Iyer, Srividhya; Kim, Ha-Neui et al. (2017) The Effects of Aging and Sex Steroid Deficiency on the Murine Skeleton Are Independent and Mechanistically Distinct. J Bone Miner Res 32:560-574
Piemontese, Marilina; Xiong, Jinhu; Fujiwara, Yuko et al. (2016) Cortical bone loss caused by glucocorticoid excess requires RANKL production by osteocytes and is associated with reduced OPG expression in mice. Am J Physiol Endocrinol Metab 311:E587-93
Fujiwara, Toshifumi; Ye, Shiqiao; Castro-Gomes, Thiago et al. (2016) PLEKHM1/DEF8/RAB7 complex regulates lysosome positioning and bone homeostasis. JCI Insight 1:e86330
Fujiwara, T; Zhou, J; Ye, S et al. (2016) RNA-binding protein Musashi2 induced by RANKL is critical for osteoclast survival. Cell Death Dis 7:e2300

Showing the most recent 10 out of 162 publications