Skip to main content
The Journals of Gerontology Series A: Biological Sciences and Medical Sciences logoLink to The Journals of Gerontology Series A: Biological Sciences and Medical Sciences
. 2013 Jul 3;68(10):1197–1208. doi: 10.1093/gerona/glt079

Basic Biology of Skeletal Aging: Role of Stress Response Pathways

Maria Almeida 1,, Charles A O’Brien 1
PMCID: PMC3779633  PMID: 23825036

Abstract

Although a decline in bone formation and loss of bone mass are common features of human aging, the molecular mechanisms mediating these effects have remained unclear. Evidence from pharmacological and genetic studies in mice has provided support for a deleterious effect of oxidative stress in bone and has strengthened the idea that an increase in reactive oxygen species (ROS) with advancing age represents a pathophysiological mechanism underlying age-related bone loss. Mesenchymal stem cells and osteocytes are long-lived cells and, therefore, are more susceptible than other types of bone cells to the molecular changes caused by aging, including increased levels of ROS and decreased autophagy. However, short-lived cells like osteoblast progenitors and mature osteoblasts and osteoclasts are also affected by the altered aged environment characterized by lower levels of sex steroids, increased endogenous glucocorticoids, and higher oxidized lipids. This article reviews current knowledge on the effects of the aging process on bone, with particular emphasis on the role of ROS and autophagy in cells of the osteoblast lineage in mice.

Key Words: Autophagy, Bone, Mesenchymal stem cells, Osteocytes, Oxidative stress.

Characteristics of the Aged Skeleton

Advancing age represents a major risk factor for the decline in bone mass and strength and, therefore, the rise in the incidence of bone fractures. Indeed, it is expected that the incidence of osteoporotic-related fractures in the United States alone will reach 3 million annually by 2025 (1). The adult skeleton is continuously remodeled throughout life by the coordinated activities of osteoclasts and osteoblasts. Osteoclasts derive from cells of the hematopoietic lineage and are responsible for bone resorption, whereas osteoblasts originate from the mesenchymal lineage and are responsible for bone formation. With aging, the amount of bone resorbed by osteoclasts is not fully restored with bone deposited by osteoblasts and this imbalance leads to loss of bone mass and strength (Table 1). The decline in whole bone strength is due to reductions in cancellous and cortical bone density, decreased cortical thickness, and a marked increase in cortical porosity (2–5). Strikingly, the age-associated increase in cortical porosity accounts for 76% of the reduction in cortical strength (6).

Table 1.

Age-Related Changes in Bone that are Common to Humans and Rodents.

Age-Related Changes in Bone Humans Rodents
Morphologic changes
 Decreased BMD (7–9) (10–12)
 Decreased cancellous bone (2) (10,13,14)
 Decreased cortical bone (2) (15–17)
 Increased cortical porosity (3–6) (15,16)
 Decreased wall width (18,19) (10)
Cellular changes
 Decreased remodeling in cancellous bone (20) (10,21)
 Increased remodeling in cortical bone (3) (22)
 Increased Ob/Ot apoptosis (23,24) (10)
 Decreased osteocyte density (23,25,26)
 Increased marrow adiposity (27–29) (30)
 Dysfunctional MSCs (31) (32,33)
Molecular changes
 Increased oxidative stress (34)
 Decreased Wnt signaling (17,34)
 Increased PPARγ (30,35)
 Decreased growth factors (36–38) (39)
 Decreased/increased RANKL (40) (21)
 Decreased/increased Sost (40) (21)

Notes: BMD = bone mineral density; Ob/Ot = osteoblast and osteocyte; PPAR = peroxisome proliferator-activated receptor; RANKL = receptor activator of nuclear factor kappa-B ligand.

The most common histological finding in aged human bone is reduced wall width—an index of the reduced amount of work (deposition of bone matrix) performed by teams of osteoblasts during bone remodeling (18,19). This deficient bone formation is due primarily to an insufficient number of osteoblasts (Table 1) (41). The defective osteoblast number in the aging skeleton has been attributed to a decrease in the number of mesenchymal stem cells (MSCs), defective proliferation and differentiation of progenitor cells, or diversion of these progenitors toward the adipocyte lineage, as well as to increased apoptosis (Figure 1). Another common histologic feature of aged human bone, which may be related to the reduced osteoblast number, is a decrease in osteocytes density (25,26). Mineralization of osteocyte lacunae, a phenomenon termed micropetrosis, may also contribute to the decrease in osteocytes density with age (23). The conditions that cause micropetrosis are unclear, but this process may be one potential outcome of osteocyte death.

Figure 1.

Figure 1.

Mechanisms of age-related changes in bone. Both cell intrinsic and cell extrinsic mechanisms contribute to the reduced osteoblast generation and increased osteoblast and osteocytes apoptosis that lead to the loss of bone mass with aging. For example, an increase in ROS and endogenous hyperglucocorticoidism with age, as well as sex steroid deficiency, promote osteoblast and osteocyte apoptosis. Osteocyte apoptosis leads to a reduction in osteocytes density.

Similar to humans, loss of bone mass and strength with advancing age in rodents is associated with an increase in the prevalence of apoptotic osteoblasts and osteocytes and a corresponding decrease in osteoblast number, and bone formation rate (10–12,42,43). Increased intracortical porosity, associated with intense remodeling activity, is also a feature of murine bone aging (15,16,22). In contrast, the number of osteoclasts in cancellous bone is diminished with age in the mouse, in line with a concomitant decrease in receptor activator of nuclear factor kappa-B ligand (RANKL) levels in the bone marrow plasma (10,21). Thus, aging seems to exert opposite effects on bone remodeling in the cancellous versus the cortical bone compartments.

The imbalance between bone resorption and formation with age is due to multiple factors, which include extrinsic and intrinsic mechanisms of cell dysfunction. This article reviews current knowledge on the mechanisms of age-related bone loss with emphasis on the role of reactive oxygen species (ROS) and autophagy.

Which Bone Cells Age?

Although there is no universally accepted definition, cellular aging is frequently described as a decline in function due to the accumulation of damage to lipids, proteins, and DNA (88). This definition then leads to the question of which bone cells experience declines in function that mediate the age-associated changes to the skeleton. Mature osteoblasts and osteoclasts responsible for the synthesis and resorption of bone matrix, respectively, are terminally differentiated postmitotic cells with a short life span (19,44). For bone formation to continue uninterrupted throughout life, osteoblasts need to be constantly replaced with new ones (45) originating from MSCs and produced through the replication and differentiation of a lineage-committed progenitor cell (Figure 1). MSCs and osteocytes—former osteoblasts buried in the bone matrix—are long-lived cells and, consequently, are likely more prone to suffer the intrinsic damaging effects of aging.

Mesenchymal Stem Cells

The loss of regenerative capacity of different tissues with aging, which in turn leads to an impaired response to stress, has led to the idea that aging is due, at least in part, to the loss of functional adult stem cells needed for tissue repair (46,47). In support of this hypothesis, old mice have a significant reduction in the number, proliferative capacity, or differentiation potential of distinct stem cells including neuronal (48,49), germline (50,51), hematopoietic (52), and muscle stem cells (53,54).

MSCs that give rise to the osteoblast lineage are commonly defined by their in vitro ability to differentiate into osteoblasts, chondrocytes, and adipocytes (55,56). These cells are found in the bone marrow and in the perivascular niche in multiple human organs (57). However, the identity and location of adult MSCs in vivo have remained elusive due to lack of specific cellular markers. Lineage tracing studies in mice have identified subsets of mesenchymal cells in the bone marrow that express smooth muscle α-actin and/or myxovirus resistance-1 that can give rise to osteoblasts present on the bone surfaces (45,58). Because these cells also persist in the marrow, these markers may identify bona fide stem cells.

Changes in the behavior of bone marrow–derived MSCs with aging have been reported in humans and rodents. These include loss of potential to proliferate and differentiate, loss of capacity to form bone in vivo, and increased senescence (31–33,59). Similar to the MSCs from bone marrow, multipotent cells from adipose tissue show an age-dependent loss of self-renewal capacity as well as an increased propensity for adipogenesis (60). In addition, MSCs from patients with Hutchinson–Gilford progeria syndrome, a disease characterized by accelerated aging, exhibit defective ability to differentiate (61).

The loss of mesenchymal progenitor functionality with age might be due to the accumulation of oxidative damage. Indeed, progenitor cells from old rats exhibit higher levels of oxidized proteins and lipids and showed decreased levels of antioxidant enzyme activity compared with cells from young rats (62). The increased oxidative stress was associated with decreased colony-forming unit fibroblast numbers, increased levels of apoptosis, and reduced proliferation and potential for differentiation. Oxidative stress is also a critical determinant of hematopoietic and neuronal stem cell dysfunction (63,64). These studies suggest a mechanistic link between intracellular oxidants and the decline in regenerative function that occurs as a normal consequence of aging.

Importantly, recent evidence has established that adult stem/progenitor cell dysfunction contributes to aging-related degeneration. Indeed, administration of muscle-derived stem/progenitor cells, isolated from young wild-type mice, to a mouse model of a human progeria conferred significant life-span and health-span extension (65). It remains unknown, however, whether the changes in adult MSCs with aging are functionally related to skeletal involution.

Osteocytes

Within cancellous bone, a subset of osteoblasts becomes embedded in bone matrix with each remodeling cycle thereby generating a fresh population of osteocytes within the new packet of bone. This process also occurs within remodeling cortical bone, but osteocytes near the periosteum are derived from the osteoblasts that continuously expand the periosteum via modeling. Regardless of their origin, all osteocytes are only as old as the bone matrix in which they are embedded, which depends on the rate of remodeling at that site. For example, osteocytes within auditory ossicles, which remodel very little after birth, are essentially as old as the individual (66). Thus, depending on their anatomical location, osteocyte life span can range from a few months to several decades.

Osteocytes have many important functions, including the control of bone resorption and formation, as well as the regulation of phosphate homeostasis. Many of these functions were only recently identified as a result of the development of genetic tools to manipulate gene expression specifically in osteocytes (67,68). For example, osteocyte-specific gene deletion studies have revealed that osteocytes control bone resorption via production of the osteoclastogenic cytokine RANKL (69,70), whereas they control phosphate homeostasis via production of Dmp1 and FGF23 (71). In addition, a combination of mouse and human genetic studies has revealed that osteocytes control bone formation via production of the Wnt-antagonist sclerostin, which is a product of the Sost gene (72–74).

Because osteocytes are long-lived cells, they are likely to be relatively more susceptible than osteoblasts or osteoclasts to the molecular changes caused by aging. Consistent with this idea, accumulation of the age-associated pigment lipofuscin has been demonstrated in osteocytes in rodent bone (75). Nonetheless, there has been little quantitative measurement of molecular damage in aged osteocytes, likely due to the difficulty in isolating pure populations of this cell type. Despite the paucity of quantitative evidence of molecular damage, extensive evidence has shown that osteocyte survival and number decline with age in rodents and humans (10,23,24). Based on the observation that osteocytes control bone resorption via production of RANKL and bone formation via production of sclerostin, one might anticipate that reduced osteocyte number might lead to altered production of these factors and consequent changes in bone remodeling. Consistent with this idea, recent studies by Halloran and colleagues have revealed that the levels of soluble RANKL and sclerostin decline with age in bone marrow supernatants from C57BL/6 mice (21). Although the change in soluble RANKL levels are consistent with reduced osteoclast number on cancellous bone, the low sclerostin levels would be expected to promote Wnt signaling and thereby osteoblast formation; yet osteoblast number is reduced with age. Thus, altered production of known osteocyte products does not, by itself, provide a clear explanation for the reduced cancellous bone turnover associated with aging. It is possible that oxidative stress together with other age-intrinsic mechanisms may alter bone remodeling by decreasing not only the number of osteocytes but also the synthetic capacity of osteocytes. Why these mechanisms have an overall different effect on the rate of remodeling in cancellous versus cortical bone remains unclear.

Cell Intrinsic Mechanisms of Skeletal Aging

Oxidative Stress

Free radical damage has been considered a key component in the tissue degeneration associated with aging (76,77). The majority of cellular ROS is generated by the mitochondria during oxidative phosphorylation. In addition, ROS can be produced in other cellular compartments by nicotinamide adenine dinucleotide phosphate oxidases, lipoxygenases, and other enzymes in response to growth factors and cytokines (78). To prevent excessive ROS production, cells scavenge ROS by multiple mechanisms including production of enzymes such as superoxide dismutases (SODs) and catalase as well as thiol-containing oligopeptides with redox-active sulfhydryl moieties, the most abundant of which are glutathione and thioredoxin (79). Several transcription factors, including FoxOs and p53, have been identified as important defense mechanism against oxidative stress. Indeed, FoxOs promote the maintenance of hematopoietic and neuronal stem cells by attenuating ROS (63,64). Growth factors inhibit FoxO activity by promoting Akt-mediated phosphorylation of FoxO1, 3, and 4, which results in their nuclear export (80–84). In contrast, growth factor depletion or increased ROS levels activate FoxOs and thereby the transcription of genes involved in free radical scavenging, cell cycle, DNA repair, and life span (85,86).

The accumulation of dysfunctional mitochondrial and a decline of antioxidant defense mechanisms with age result in an increase in the levels of free radicals. A sustained excess of ROS damages proteins, lipids, and DNA (87,88). Indeed, oxidative stress is responsible for age-related tissue damage and various disease states such as diabetes, cardiovascular diseases, cancer, and neurodegeneration (89).

Oxidative stress also increases in bone with age (10,17). Evidence from pharmacological and genetic studies in mice has provided support for a deleterious effect of oxidative stress in bone. Specifically, the loss of bone caused by gonadectomy in males or females is attenuated by antioxidants (10,90,91). In contrast, mice treated with the prooxidant buthionine sulfoximine and murine models of premature aging associated with oxidative damage exhibit low bone mass (91–93). Importantly, mice with global deletion of the antioxidant gene Sod1 exhibit low bone mass, which worseness with age (94). Similar to the aged skeleton of wild-type mice, Sod1 /− mice exhibit decreased trabecular and cortical bone mass, which is associated with lower osteoblast and osteoclast numbers. Administration of an antioxidant to Sod1 /− mice normalizes these defects implicating ROS as the main cause of the low bone mass in this model.

Further support for a role of oxidative stress in skeletal homeostasis was provided by murine models of FoxO loss or gain of function. Specifically, conditional deletion of FoxO1, 3, and 4 in young mice or targeted deletion of FoxO1 in osteoblasts resulted in an increase in oxidative stress in bone, a decrease in the number of osteoblasts, and low bone mass (95,96). Conversely, overexpression of a FoxO3 transgene in mature osteoblasts decreased oxidative stress and increased osteoblast number and bone mass.

At the cellular level, oxidative stress decreases the life span of osteoblast in bone, as highlighted by the observation that administration of antioxidants abrogates osteoblast apoptosis in ovariectomized or aged mice (10,17). In addition, osteoblasts from Sod1- or FoxO-null mice exhibit decreased life span (94,95).

ROS also inhibit the Wnt/β-catenin signaling pathway. β-Catenin is indispensable for osteoblastogenesis during development and in adulthood, and loss or gain of function of this pathway is associated with a pronounced decrease or increase of bone mass, respectively, in humans and mice (97). Wnt proteins bind to the Frizzled/LRP5 or LRP6 receptor complex, thereby preventing the proteasomal degradation of the transcriptional coactivator β-catenin (98). β-Catenin translocates into the nucleus where it associates with the T-cell factor (TCF) lymphoid-enhancer binding factor family of transcription factors and regulates the expression of Wnt target genes. In the setting of oxidative stress or nutrient depletion, FoxOs divert the limited pool of active β-catenin from TCF- to FoxO-mediated transcription in diverse cell types, including osteoblasts, colon cancer cells, and hepatocytes (34,99,100). In line with this, targeted deletion of FoxOs in osteoblast progenitors has elucidated that FoxOs restrain the pro-proliferative effects of Wnt signaling on these cells and thereby attenuate bone formation throughout life (101). Similar to the osteoblast progenitors, deletion of FoxOs in enteroendocrine progenitors or in neuronal progenitors increases β-catenin/TCF-mediated transcription and proliferation (64,102). Thus, diversion of β-catenin from TCF- to FoxO-mediated transcription in response to stressful conditions, such as the increased ROS and growth factor deficiency that occur with aging (Table 1), may represent a pathogenetic mechanism for osteoporosis and perhaps several other degenerative diseases associated with old age (103–107).

Autophagy

Autophagy is a process in which cellular components, such as long-lived proteins and organelles, are degraded by the lysosome to maintain the health and viability of the cell. Under normal physiological conditions, autophagy acts as a quality control system that removes defective organelles or protein aggregates. However, during stressful conditions, such as nutrient deprivation or hypoxia, autophagy is increased to break down cellular components for use as an energy source. There are several forms of autophagy including chaperone-mediated autophagy, microautophagy, and macroautophagy (108). Chaperone-mediated autophagy involves the targeting of proteins containing a specific five amino acid motif directly to the lysosome for degradation, whereas microautophagy involves invagination of the lysosomal membrane to engulf small portions of the cytoplasm. Macroautophagy refers to a process whereby large components of the cytoplasm, including mitochondria and protein aggregates, are surrounded by a double-membrane structure to form a vacuole known as an autophagosome, which eventually fuses with the lysosome to allow degradation of the components (109) (Figure 2). The most prevalent form of autophagy in many cell types is macroautophagy and hereafter we will use the general term autophagy to refer to this process.

Figure 2.

Figure 2.

Autophagy degrades and recycles cellular components. Autophagy is an intracellular recycling pathway in which cellular components, including protein aggregates and organelles such as mitochondria, are targeted to the lysosome for degradation. Cellular components targeted for degradation are engulfed by a double-membrane structure known an autophagosome. Autophagosome formation depends on series of ubiquitin-like conjugation reactions. In this process, Atg7, which is an E1-like enzyme, activates a ubiquitin-like protein known as LC3. LC3 then becomes conjugated to phosphatidyl ethanolamine (PE) and thereby promotes autophagosome production. Importantly, Atg7 is essential for autophagy and conditional deletion of this gene can be used to examine the importance of autophagy in specific cell types.

After initiation of the autophagy process, autophagosome formation is controlled in part by activation of a ubiquitin-like protein known as LC3 (110). LC3 is activated by the E1-like enzyme Atg7 and is eventually conjugated to phosphatidyl ethanolamine in the growing double-membrane structure. It is important to note that this conjugation process is absolutely dependent on Atg7 and that genetic inactivation of Atg7 effectively suppresses autophagy (111). Thus, conditional deletion of Atg7 is frequently used to determine the role of autophagy in a particular cell type or tissue.

Because aging is associated with the accumulation of damaged cellular components, it has been proposed that autophagy may decrease or become less efficient with age (112–117). Consistent with this idea, autophagy in the kidney declines in aged mice and can be restored by caloric restriction (118). Similarly, expression of some autophagy-related genes declines in brain tissue from aged humans and in chondrocytes in osteoarthritic bones from aged mice (119,120).

Additional work suggests that autophagy contributes to the long-term health of cells and organisms. Specifically, an intact autophagy system is required for the life-span-extending action of caloric restriction or daf-2 pathway mutations in Caenorhabditis elegans (114) and promotion of autophagy extends life span in Drosophila (121). Moreover, deletion of autophagy genes in myocytes, pancreatic β cells, or T cells leads to accumulation of damaged mitochondria and increased ROS production (122–127). These latter studies suggest the possibility that age-associated cellular damage is due in part to a decline in autophagy, leading to damaged mitochondria, which in turn produce more ROS. Elevated ROS would then be expected to perpetuate the cycle of damage. Another possibility is that ROS is elevated with age, independent of changes in autophagy, and this leads to damaged mitochondria, which are not replaced effectively due to reduced levels of autophagy in aged cells. The accumulation of damaged mitochondria would then be expected to accentuate ROS production (128).

Because osteocytes are long-lived postmitotic cells that can only be replaced by bone turnover, it is reasonable to hypothesize that autophagy plays an important role in their survival and function. Specifically, autophagy may help osteocytes defend against stresses such as elevated ROS. To address the role of autophagy in osteocytes, we have deleted Atg7 in osteocytes using the dentin matrix protein 1 (Dmp1)-Cre transgenic mouse developed by Feng and colleagues (68). Preliminary analyses of these mice revealed that they have low bone mass at 6 months of age that is associated with low bone remodeling (129). The magnitude of these changes was similar to that observed when comparing old (>18 months old) and young (<8 months old) mice (10). Importantly, oxidative stress was elevated in the bones of mice lacking autophagy in osteocytes, as measured by ROS production in bone marrow cells and by p66shc phosphorylation in bone (130). Thus, suppression of autophagy in osteocytes was sufficient to cause skeletal changes in young adult mice that are similar to those observed in aged wild-type mice. Additional studies will be required to identify the molecular mechanisms by which autophagy in osteocytes controls bone remodeling and bone mass. Perhaps more importantly, it will be important to determine whether autophagy does indeed decline with age in osteocytes or cells at any stage of osteoblast differentiation.

Recent evidence suggests that autophagy may also be important for the long-term health of progenitors as well as fully differentiated long-lived cell types. Specifically, deletion of Atg7 from hematopoietic stem cells (HSCs) in mice led to accumulation of mitochondria and increased oxidative stress in the HSCs, which was associated with increased proliferation and DNA damage (131). These results led to the conclusion that autophagy is required for maintenance of the HSC compartment in adult mice. Another study has shown that the basal level of autophagy is elevated in HSCs of old mice and is required for the survival of these cells under stressful conditions, such as nutrient deprivation (132). Importantly, FoxO3 is critical for expression of autophagy genes and the induction of autophagy in response to stress in HSCs. Thus, genes encoding the machinery for the autophagic response are important targets for FoxOs in HSCs and other cell types. It is possible that autophagy may play a similar role in the maintenance of MSCs in bone and that changes in autophagy in this compartment may underlie changes in MSC behavior with age.

Cell Extrinsic Mechanisms of Skeletal Aging

Loss of Sex Steroids

In women, the loss of bone occurs at a faster rate after the menopause, attesting to the adverse role of estrogen deficiency on bone mass and its contribution to the acceleration of skeletal involution with age. Estrogen deficiency causes an increase in bone remodeling, increased osteoclastogenesis and osteoblastogenesis, increased osteoclast and osteoblast numbers, and increased resorption and formation—albeit unbalanced. Conversely, estrogens slow the rate of bone remodeling and promote a positive balance between bone formation and resorption by attenuating the generation of osteoclast and osteoblast progenitors in the bone marrow and exerting a proapoptotic effect on osteoclasts and an antiapoptotic effect on osteoblasts and osteocytes (41,43,133). Estrogens decrease osteoclast generation and life span via direct effects on osteoclasts mediated by the estrogen receptor α (134,135). These actions are responsible for the protective effects of estrogens on the cancellous bone compartment. On the other hand, the estrogen receptor α present in osteoblast progenitors mediates a protective effect of estrogens against endocortical bone resorption (136). Attenuation of the production of cytokines like IL6, IL1, TNFα, and RANKL by other cells present in the bone marrow environment, including T- and B lymphocytes and stromal/osteoblastic cells, also contribute to the antiosteoclastogenic properties of estrogens (137,138).

Like aging, gonadectomy in female or male mice promotes an increase in oxidative stress in bone (43). Administration of antioxidants, similar to estrogens or androgens, abrogates the stimulatory effects of gonadectomy on oxidative stress, osteoclastogenesis, and osteoblast and osteocytes apoptosis, and it prevents the loss of bone mass (10,90). Hence, sex steroid deficiency may contribute to age-related bone loss, at least in part, by increasing oxidative stress.

Relative Excess of Endogenous Glucocorticoids

Administration of glucocorticoids is a common cause of osteoporosis (139). Glucocorticoids are strong inhibitors of bone formation, at least in part, by stimulating osteoblast apoptosis. ROS generation and activation of a PKCβ/p66shc/JNK signaling cascade are responsible for the proapoptotic effects of glucocorticoids on osteoblastic cells (140). Moreover, glucocorticoids promote osteocyte apoptosis via activation of Pyk2 and JNK, followed by inside-out signaling that leads to cell detachment–induced apoptosis or anoikis (141).

Glucocorticoids also suppress the generation of new osteoblasts by attenuating Wnt signaling. This occurs via at least three separate mechanisms: first, glucocorticoids stimulate expression of the Wnt inhibitor DKK1 in bone and osteoblastic cell cultures (142); second, glucocorticoids suppress the PI3K/Akt/GSK3β signaling pathway (143); and third, suppression of Akt leads to activation of FoxO transcription factors, which antagonize Wnt/β-catenin signaling (140).

Glucocorticoids also modulate bone resorption and increase cortical porosity. The rapid loss of bone caused by glucocorticoid excess results from direct prosurvival actions on osteoclasts (144). The short-term transient increase in bone resorption in combination with long-term impairment of bone formation might be responsible for the increased cortical porosity in patients treated long term with glucocorticoids (145).

Importantly, the production of endogenous glucocorticoids as well as the sensitivity of bone cells to glucocorticoids increases with age (146,147). In line with this evidence, mice with osteoblast/osteocyte-specific transgenic expression of 11β-HSD2, the enzyme that inactivates glucocorticoids, are partially protected from the adverse effects of aging on osteoblast and osteocyte apoptosis, bone formation rate, and bone strength (147). Thus, the rise in endogenous glucocorticoids represents another age-associated pathogenetic mechanism of involutional osteoporosis.

Marrow Adipogenesis and Lipid Oxidation

With advancing age, the number of adipocytes in the bone marrow increases dramatically, particularly in long bones, in both humans and rodents. In line with the changes in adiposity, the expression of peroxisome proliferator-activated receptor (PPAR) γ2, a transcription factor that is essential for adipogenesis, is increased with age in the bone of mice (30,35). Osteoblasts and bone marrow adipocytes arise from a common progenitor cell and lineage allocation into adipocytes or osteoblasts is considered to be reciprocally exclusive (Figure 1). In view of these findings, it has been proposed that enhanced PPARγ2 expression in progenitor cells plays an important role in the pathogenesis of age-related bone loss by favoring the commitment of mesenchymal progenitors to the adipocyte instead of the osteoblast lineage. In support of this hypothesis is the evidence that mice haploinsufficient for PPARγ2 exhibit increased osteoblast number and high bone mass (148).

Entry of mesenchymal progenitor cells into the osteoblastic lineage is dependent on Wnt/β-catenin signaling. In addition to its role in osteoblastogenesis, Wnt signaling is a strong inhibitor of adipogenesis. Wnt/β-catenin signaling inhibits adipogenesis, at least in part, by suppressing PPARγ expression (149,150). In agreement with this, deletion of β-catenin in osteoprogenitor cells in adult mice increases the expression of PPARγ2 and bone marrow adiposity and decreases bone mass (151). On the other hand, activation of PPARγ by ligands such as oxidized polyunsaturated fatty acids promotes PPARγ association with β-catenin and induces β-catenin degradation (30,152), thereby decreasing β-catenin/TCF-mediated transcription. The inverse relationship between adipocyte and osteoblast differentiation notwithstanding, further studies are needed to demonstrate a causal role of the increased marrow adipogenesis to the decrease in bone mass with aging.

Lipid oxidation plays a critical role in the development of atherogenesis and increases with age in bone. In the process of lipid oxidation, lipoxygenases, such as Alox15, oxidize polyunsaturated fatty acids to form products that bind to and activate PPARγ and generate prooxidants like 4-HNE (153). Mice lacking Alox15 exhibit increased bone mass (154), suggesting that lipid oxidation exerts a deleterious effect on bone. In line with this, both 4-HNE, via FoxO activation, and oxidized polyunsaturated fatty acids, via PPARγ, attenuate β-catenin/TCF-mediated transcription and osteoblast generation (30). Oxidized lipids also stimulate apoptosis of osteoblastic cells and inhibit BMP-2-induced osteoblast differentiation via ROS-independent mechanisms (155–157). Thus, lipid oxidation potentiates oxidative stress and may contribute to the reduction in bone formation that occurs with aging.

Decreased Growth Factors

Growth factors like growth hormone and insulin-like growth factor 1 (IGF-1) are critical for skeletal growth, and IGF-1 is the most abundant growth factor deposited in the bone matrix throughout life (158). The bioactivity of IGF-1 is regulated by the interaction with IGF-binding proteins (IGFBP-1–6) (159). Clinical studies along with murine models have suggested that physiological levels of IGFBPs contribute to the attainment and maintenance of optimal bone mass, at least in part, by targeting IGF-1 to the skeleton (160). IGF-1 promotes the differentiation of osteoprogenitor cells through activation of a PI3K/Akt/mTOR pathway (39,161). In line with this, deletion of the IGF-1 receptor in osteoblasts decreases bone formation rate and cancellous bone volume (39,162). In addition, IGF-1 is the primary osteogenic factor released from the bone matrix via the resorptive action of osteoclast.

In humans, the levels of IGF-1 in the circulation and bone matrix decline substantially with age, most likely due to a reduction in the secretion of growth hormone. Indeed, between 20 and 60 years of age, the content of IGF-1 in human bones declines by 60% (163). Importantly, the levels of IGF-1 and IGFBP3 in the bone matrix are also correlated with the decrease in bone mineral density with aging and with increases in the total number of hip fractures (164,165). Similar to humans, the reduction in the levels of IGF-1 in bone marrow and matrix from 8 to 20 months of age in rats is associated with the decline in bone mass (39). Notably, injections of IGF-1 combined with IGFBP3, the predominant IGFBP, in the long bones of aged rats stimulates bone formation and increases cancellous bone mass. It is, therefore, possible that a decrease in IGF-1 released from the bone matrix, most likely due to a reduction in IGF-1 content along with decreased resorption, contributes to the age-associated decline in bone mass.

Other Age-Related Changes

Chronic inflammation and reduced physical activity are additional extrinsic factors that are often associated with human aging and potentially mediate some of the negative impacts of aging on the skeleton (166,167). Although inflammatory cytokines clearly play an important role in the bone loss caused by sex steroid deficiency, it is less clear that they are involved in age-associated bone loss, at least in rodents. Specifically, inflammatory bone loss is associated with elevated osteoclast formation in cancellous bone, whereas osteoclast number is low in the cancellous bone of aged mice compared with young mice (10,168). Thus the histological picture of aged bone is not consistent with a role for chronic inflammation. Similarly, biomechanical unloading causes bone loss, at least in part, via increased resorption. Thus, the histological changes that occur in the aged skeleton also do not appear to be caused by unloading. Be that as it may, it remains likely that chronic inflammation and reduced physical activity do contribute to osteoporosis in certain individuals, even though these mechanisms may not be generally involved.

Conclusions

Although a decline in bone formation and loss of bone mass are common features of human aging, the molecular mechanisms mediating these effects have remained unclear. Evidence from pharmacological and genetic studies in mice has provided support for a deleterious effect of oxidative stress in bone and has strengthened the idea that an increase in ROS with advancing age represents a pathophysiological mechanism for age-related osteoporosis. The contribution of a decline in mitochondrial function and an increase in ROS to age-related diseases and life span notwithstanding, other mechanisms including deregulated autophagy, telomere shortening, and abnormal protein folding are likely to represent additional critical effectors of aging. Furthermore, several cell extrinsic mechanisms, including a decline in sex steroids, an increase in endogenous glucocorticoids, and higher lipid oxidation are associated with the decreased bone formation seen in old age (Figure 2). Thus, the cellular changes that occur in the skeleton with advancing age appear to result from numerous independent mechanisms, although elevated oxidative stress may be a common component of many of them. It will now be important to determine whether suppression of oxidative stress in specific cell populations can ameliorate any aspects of skeletal aging.

Although the long-lived cells in bone, such as the adult MSCs and osteocytes, are more prone to suffer the negative effects of aging, short-lived cells like osteoblast progenitors and mature osteoblasts are also affected by the altered aged environment. Understanding how different anti- or pro-aging molecules exert their effects at different stages of osteoblast differentiation will be challenging but crucial for the elucidation of the signaling networks operating in aged bone. Deciphering such ageing networks, together with identification of the cells most affected by the ageing mechanisms, should provide the information necessary for the advancement and development of therapeutic strategies to prevent or treat age-related osteoporosis.

Funding

This work was supported by the National Institutes of Health (R01 AR56679, P01 AG13918, and R01 AR49794), the Department of Veterans Affairs from the Biomedical Laboratory Research and Development Service of the VA Office of Research and Development to C.A.O. (5I01BX000294), the University of Arkansas for Medical Sciences (UAMS) Translational Research Institute (UL1 RR029884), and Tobacco Settlement funds.

References

  • 1. Burge R, Dawson-Hughes B, Solomon DH, Wong JB, King A, Tosteson A. Incidence and economic burden of osteoporosis-related fractures in the United States, 2005–2025. J Bone Miner Res. 2007;22:465–475 [DOI] [PubMed] [Google Scholar]
  • 2. Riggs BL, Melton LJ, Robb RA, et al. A population-based assessment of rates of bone loss at multiple skeletal sites: evidence for substantial trabecular bone loss in young adult women and men. J Bone Miner Res. 2008;23:205–214 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Zebaze RM, Ghasem-Zadeh A, Bohte A, et al. Intracortical remodelling and porosity in the distal radius and post-mortem femurs of women: a cross-sectional study. Lancet. 2010;375:1729–1736 [DOI] [PubMed] [Google Scholar]
  • 4. Parfitt AM. Age-related structural changes in trabecular and cortical bone: cellular mechanisms and biomechanical consequences. Calcif Tissue Int. 1984;36(suppl 1):S123–S128 [DOI] [PubMed] [Google Scholar]
  • 5. Burghardt AJ, Kazakia GJ, Ramachandran S, Link TM, Majumdar S. Age- and gender-related differences in the geometric properties and biomechanical significance of intracortical porosity in the distal radius and tibia. J Bone Miner Res. 2010;25:983–993 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. McCalden RW, McGeough JA, Barker MB, Court-Brown CM. Age-related changes in the tensile properties of cortical bone. The relative importance of changes in porosity, mineralization, and microstructure. J Bone Joint Surg Am. 1993;75:1193–1205 [DOI] [PubMed] [Google Scholar]
  • 7. Looker AC, Wahner HW, Dunn WL, et al. Updated data on proximal femur bone mineral levels of US adults. Osteoporos Int. 1998;8:468–489 [DOI] [PubMed] [Google Scholar]
  • 8. Khosla S, Riggs BL. Pathophysiology of age-related bone loss and osteoporosis. Endocrinol Metab Clin North Am. 2005;34:1015–1030–xi [DOI] [PubMed] [Google Scholar]
  • 9. Seeman E. Invited review: pathogenesis of osteoporosis. J Appl Physiol. 2003;95:2142–2151 [DOI] [PubMed] [Google Scholar]
  • 10. Almeida M, Han L, Martin-Millan M, et al. Skeletal involution by age-associated oxidative stress and its acceleration by loss of sex steroids. J Biol Chem. 2007;282:27285–27297 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Halloran BP, Ferguson VL, Simske SJ, Burghardt A, Venton LL, Majumdar S. Changes in bone structure and mass with advancing age in the male C57BL/6J mouse. J Bone Miner Res. 2002;17: 1044–1050 [DOI] [PubMed] [Google Scholar]
  • 12. Iida H, Fukuda S. Age-related changes in bone mineral density, cross-sectional area and strength at different skeletal sites in male rats. J Vet Med Sci. 2002;64:29–34 [DOI] [PubMed] [Google Scholar]
  • 13. Bar-Shira-Maymon B, Coleman R, Cohen A, Steinhagen-Thiessen E, Silbermann M. Age-related bone loss in lumbar vertebrae of CW-1 female mice: a histomorphometric study. Calcif Tissue Int. 1989;44:36–45 [DOI] [PubMed] [Google Scholar]
  • 14. Weiss A, Arbell I, Steinhagen-Thiessen E, Silbermann M. Structural changes in aging bone: osteopenia in the proximal femurs of female mice. Bone. 1991;12:165–172 [DOI] [PubMed] [Google Scholar]
  • 15. Silbermann M, Weiss A, Reznick AZ, Eilam Y, Szydel N, Gershon D. Age-related trend for osteopenia in femurs of female C57BL/6 mice. Compr Gerontol A. 1987;1:45–51 [PubMed] [Google Scholar]
  • 16. Ferguson VL, Ayers RA, Bateman TA, Simske SJ. Bone development and age-related bone loss in male C57BL/6J mice. Bone. 2003;33:387–398 [DOI] [PubMed] [Google Scholar]
  • 17. Jilka RL, Almeida M, Ambrogini E, et al. Decreased oxidative stress and greater bone anabolism in the aged, as compared to the young, murine skeleton by parathyroid hormone. Aging Cell. 2010;9:851–867 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Lips P, Courpron P, Meunier PJ. Mean wall thickness of trabecular bone packets in the human iliac crest: changes with age. Calcif Tissue Res. 1978;26:13–17 [DOI] [PubMed] [Google Scholar]
  • 19. Parfitt AM. Bone-forming cells in clinical conditions. In: Hall BK, ed. Bone. Volume 1. The Osteoblast and Osteocyte. Boca Raton, FL: Telford Press and CRC Press; 1990:351–429 [Google Scholar]
  • 20. Parfitt AM, Villanueva AR, Foldes J, Rao DS. Relations between histologic indices of bone formation: implications for the pathogenesis of spinal osteoporosis. J Bone Miner Res. 1995;10:466–473 [DOI] [PubMed] [Google Scholar]
  • 21. Shahnazari M, Dwyer D, Chu V, et al. Bone turnover markers in peripheral blood and marrow plasma reflect trabecular bone loss but not endocortical expansion in aging mice. Bone. 2012;50:628–637 [DOI] [PubMed] [Google Scholar]
  • 22. Jilka R, Deloose A, Climer L, et al. Dysfunctional osteocytes increase RANKL and promote cortical pore formation in their vicinity: a mechanistic explanation for the development of cortical porosity with age. J Bone Miner Res. 2012;27:S348 (abstract) [Google Scholar]
  • 23. Busse B, Djonic D, Milovanovic P, et al. Decrease in the osteocyte lacunar density accompanied by hypermineralized lacunar occlusion reveals failure and delay of remodeling in aged human bone. Aging Cell. 2010;9:1065–1075 [DOI] [PubMed] [Google Scholar]
  • 24. Qiu S, Rao DS, Palnitkar S, Parfitt AM. Age and distance from the surface but not menopause reduce osteocyte density in human cancellous bone. Bone. 2002;31:313–318 [DOI] [PubMed] [Google Scholar]
  • 25. Qiu S, Rao DS, Palnitkar S, Parfitt AM. Relationships between osteocyte density and bone formation rate in human cancellous bone. Bone. 2002;31:709–711 [DOI] [PubMed] [Google Scholar]
  • 26. Manolagas SC, Parfitt AM. What old means to bone. Trends Endocrinol Metab. 2010;21:369–374 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Meunier P, Aaron J, Edouard C, Vignon G. Osteoporosis and the replacement of cell populations of the marrow by adipose tissue. A quantitative study of 84 iliac bone biopsies. Clin Orthop Relat Res. 1971;80:147–154 [DOI] [PubMed] [Google Scholar]
  • 28. Verma S, Rajaratnam JH, Denton J, Hoyland JA, Byers RJ. Adipocytic proportion of bone marrow is inversely related to bone formation in osteoporosis. J Clin Pathol. 2002;55:693–698 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Wehrli FW, Hopkins JA, Hwang SN, Song HK, Snyder PJ, Haddad JG. Cross-sectional study of osteopenia with quantitative MR imaging and bone densitometry. Radiology. 2000;217:527–538 [DOI] [PubMed] [Google Scholar]
  • 30. Almeida M, Ambrogini E, Han L, Manolagas SC, Jilka RL. Increased lipid oxidation causes oxidative stress, increased peroxisome proliferator-activated receptor-gamma expression, and diminished pro-osteogenic Wnt signaling in the skeleton. J Biol Chem. 2009;284:27438–27448 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Stenderup K, Justesen J, Clausen C, Kassem M. Aging is associated with decreased maximal life span and accelerated senescence of bone marrow stromal cells. Bone. 2003;33:919–926 [DOI] [PubMed] [Google Scholar]
  • 32. Sethe S, Scutt A, Stolzing A. Aging of mesenchymal stem cells. Ageing Res Rev. 2006;5:91–116 [DOI] [PubMed] [Google Scholar]
  • 33. Kasper G, Mao L, Geissler S, et al. Insights into mesenchymal stem cell aging: involvement of antioxidant defense and actin cytoskeleton. Stem Cells. 2009;27:1288–1297 [DOI] [PubMed] [Google Scholar]
  • 34. Almeida M, Han L, Martin-Millan M, O’Brien CA, Manolagas SC. Oxidative stress antagonizes Wnt signaling in osteoblast precursors by diverting beta-catenin from T cell factor- to forkhead box O-mediated transcription. J Biol Chem. 2007;282:27298–27305 [DOI] [PubMed] [Google Scholar]
  • 35. Lecka-Czernik B, Rosen CJ, Kawai M. Skeletal aging and the adipocyte program: new insights from an “old” molecule. Cell Cycle. 2010;9:3648–3654 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Boonen S, Aerssens J, Dequeker J, et al. Age-associated decline in human femoral neck cortical and trabecular content of insulin-like growth factor I: potential implications for age-related (type II) osteoporotic fracture occurrence. Calcif Tissue Int. 1997;61:173–178 [DOI] [PubMed] [Google Scholar]
  • 37. Bennett AE, Wahner HW, Riggs BL, Hintz RL. Insulin-like growth factors I and II: aging and bone density in women. J Clin Endocrinol Metab. 1984;59:701–704 [DOI] [PubMed] [Google Scholar]
  • 38. Giustina A, Veldhuis JD. Pathophysiology of the neuroregulation of growth hormone secretion in experimental animals and the human. Endocr Rev. 1998;19:717–797 [DOI] [PubMed] [Google Scholar]
  • 39. Xian L, Wu X, Pang L, et al. Matrix IGF-1 maintains bone mass by activation of mTOR in mesenchymal stem cells. Nat Med. 2012;18:1095–1101 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Khosla S. Pathogenesis of age-related bone loss in humans. J Gerontol A Biol Sci Med Sci. 2012. 10.1093/gerona/gls163 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Manolagas SC. Birth and death of bone cells: basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis. Endocr Rev. 2000;21:115–137 [DOI] [PubMed] [Google Scholar]
  • 42. Syed FA, Mödder UI, Roforth M, et al. Effects of chronic estrogen treatment on modulating age-related bone loss in female mice. J Bone Miner Res. 2010;25:2438–2446 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Manolagas SC. From estrogen-centric to aging and oxidative stress: a revised perspective of the pathogenesis of osteoporosis. Endocr Rev. 2010;31:266–300 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Weinstein RS, Jilka RL, Parfitt AM, Manolagas SC. Inhibition of osteoblastogenesis and promotion of apoptosis of osteoblasts and osteocytes by glucocorticoids: potential mechanisms of their deleterious effects on bone. J Clin Invest. 1998;102:274–282 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Park D, Spencer JA, Koh BI, et al. Endogenous bone marrow MSCs are dynamic, fate-restricted participants in bone maintenance and regeneration. Cell Stem Cell. 2012;10:259–272 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Sharpless NE, DePinho RA. How stem cells age and why this makes us grow old. Nat Rev Mol Cell Biol. 2007;8:703–713 [DOI] [PubMed] [Google Scholar]
  • 47. Rossi DJ, Jamieson CH, Weissman IL. Stems cells and the pathways to aging and cancer. Cell. 2008;132:681–696 [DOI] [PubMed] [Google Scholar]
  • 48. Kuhn HG, Dickinson-Anson H, Gage FH. Neurogenesis in the dentate gyrus of the adult rat: age-related decrease of neuronal progenitor proliferation. J Neurosci. 1996;16:2027–2033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Molofsky AV, Slutsky SG, Joseph NM, et al. Increasing p16INK4a expression decreases forebrain progenitors and neurogenesis during ageing. Nature. 2006;443:448–452 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Zhang X, Ebata KT, Robaire B, Nagano MC. Aging of male germ line stem cells in mice. Biol Reprod. 2006;74:119–124 [DOI] [PubMed] [Google Scholar]
  • 51. Ryu BY, Orwig KE, Oatley JM, Avarbock MR, Brinster RL. Effects of aging and niche microenvironment on spermatogonial stem cell self-renewal. Stem Cells. 2006;24:1505–1511 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Morrison SJ, Wandycz AM, Akashi K, Globerson A, Weissman IL. The aging of hematopoietic stem cells [see comments]. Nat Med. 1996;2:1011–1016 [DOI] [PubMed] [Google Scholar]
  • 53. Conboy IM, Conboy MJ, Smythe GM, Rando TA. Notch-mediated restoration of regenerative potential to aged muscle. Science. 2003;302:1575–1577 [DOI] [PubMed] [Google Scholar]
  • 54. Shefer G, Van de Mark DP, Richardson JB, Yablonka-Reuveni Z. Satellite-cell pool size does matter: defining the myogenic potency of aging skeletal muscle. Dev Biol. 2006;294:50–66 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Friedenstein AJ, Chailakhyan RK, Latsinik NV, Panasyuk AF, Keiliss-Borok IV. Stromal cells responsible for transferring the microenvironment of the hemopoietic tissues. Cloning in vitro and retransplantation in vivo. Transplantation. 1974;17:331–340 [DOI] [PubMed] [Google Scholar]
  • 56. Caplan AI. Mesenchymal stem cells. J Orthop Res. 1991;9:641–650 [DOI] [PubMed] [Google Scholar]
  • 57. Crisan M, Yap S, Casteilla L, et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell. 2008;3:301–313 [DOI] [PubMed] [Google Scholar]
  • 58. Grcevic D, Pejda S, Matthews BG, et al. In vivo fate mapping identifies mesenchymal progenitor cells. Stem Cells. 2012;30:187–196 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Coipeau P, Rosset P, Langonne A, et al. Impaired differentiation potential of human trabecular bone mesenchymal stromal cells from elderly patients. Cytotherapy. 2009;11:584–594 [DOI] [PubMed] [Google Scholar]
  • 60. Huang SC, Wu TC, Yu HC, et al. Mechanical strain modulates age-related changes in the proliferation and differentiation of mouse adipose-derived stromal cells. BMC Cell Biol. 2010;11:18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Scaffidi P, Misteli T. Lamin A-dependent misregulation of adult stem cells associated with accelerated ageing. Nat Cell Biol. 2008;10:452–459 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Stolzing A, Scutt A. Age-related impairment of mesenchymal progenitor cell function. Aging Cell. 2006;5:213–224 [DOI] [PubMed] [Google Scholar]
  • 63. Tothova Z, Kollipara R, Huntly BJ, et al. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell. 2007;128:325–339 [DOI] [PubMed] [Google Scholar]
  • 64. Paik JH, Ding Z, Narurkar R, et al. FoxOs cooperatively regulate diverse pathways governing neural stem cell homeostasis. Cell Stem Cell. 2009;5:540–553 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Lavasani M, Robinson AR, Lu A, et al. Muscle-derived stem/progenitor cell dysfunction limits healthspan and lifespan in a murine progeria model. Nat Commun. 2012;3:608 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Marotti G, Farneti D, Remaggi F, Tartari F. Morphometric investigation on osteocytes in human auditory ossicles. Ann Anat. 1998;180:449–453 [DOI] [PubMed] [Google Scholar]
  • 67. Bonewald LF. The amazing osteocyte. J Bone Miner Res. 2011;26:229–238 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Lu Y, Xie Y, Zhang S, Dusevich V, Bonewald LF, Feng JQ. DMP1-targeted Cre expression in odontoblasts and osteocytes. J Dent Res. 2007;86:320–325 [DOI] [PubMed] [Google Scholar]
  • 69. Xiong J, Onal M, Jilka RL, Weinstein RS, Manolagas SC, O’Brien CA. Matrix-embedded cells control osteoclast formation. Nat Med. 2011;17:1235–1241 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Nakashima T, Hayashi M, Fukunaga T, et al. Evidence for osteocyte regulation of bone homeostasis through RANKL expression. Nat Med. 2011;17:1231–1234 [DOI] [PubMed] [Google Scholar]
  • 71. Feng JQ, Ward LM, Liu S, et al. Loss of DMP1 causes rickets and osteomalacia and identifies a role for osteocytes in mineral metabolism. Nat Genet. 2006;38:1310–1315 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Brunkow ME, Gardner JC, Van Ness J, et al. Bone dysplasia sclerosteosis results from loss of the SOST gene product, a novel cystine knot-containing protein. Am J Hum Genet. 2001;68:577–589 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Balemans W, Patel N, Ebeling M, et al. Identification of a 52kb deletion downstream of the SOST gene in patients with van Buchem disease. J Med Genet. 2002;39:91–97 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Li X, Ominsky MS, Niu QT, et al. Targeted deletion of the sclerostin gene in mice results in increased bone formation and bone strength. J Bone Miner Res. 2008;23:860–869 [DOI] [PubMed] [Google Scholar]
  • 75. Tonna EA. Accumulation of lipofuscin (age pigment) in aging skeletal connective tissues as revealed by electron microscopy. J Gerontol. 1975;30:3–8 [DOI] [PubMed] [Google Scholar]
  • 76. Harman D. Aging: a theory based on free radical and radiation chemistry. J Gerontol. 1956;11:298–300 [DOI] [PubMed] [Google Scholar]
  • 77. Muller FL, Lustgarten MS, Jang Y, Richardson A, Van Remmen H. Trends in oxidative aging theories. Free Radic Biol Med. 2007;43:477–503 [DOI] [PubMed] [Google Scholar]
  • 78. Janssen-Heininger YM, Mossman BT, Heintz NH, et al. Redox-based regulation of signal transduction: principles, pitfalls, and promises. Free Radic Biol Med. 2008;45:1–17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Dickinson DA, Forman HJ. Glutathione in defense and signaling: lessons from a small thiol. Ann N Y Acad Sci. 2002;973:488–504 [DOI] [PubMed] [Google Scholar]
  • 80. Brunet A, Bonni A, Zigmond MJ, et al. Akt promotes cell survival by phosphorylating and inhibiting a forkhead transcription factor. Cell. 1999;96:857–868 [DOI] [PubMed] [Google Scholar]
  • 81. Kops GJ, de Ruiter ND, De Vries-Smits AM, Powell DR, Bos JL, Burgering BM. Direct control of the forkhead transcription factor AFX by protein kinase B. Nature. 1999;398:630–634 [DOI] [PubMed] [Google Scholar]
  • 82. Biggs WH, III, Meisenhelder J, Hunter T, Cavenee WK, Arden KC. Protein kinase B/Akt-mediated phosphorylation promotes nuclear exclusion of the winged helix transcription factor FKHR1. Proc Natl Acad Sci U S A. 1999;96:7421–7426 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Tang ED, Nuñez G, Barr FG, Guan KL. Negative regulation of the forkhead transcription factor FKHR by Akt. J Biol Chem. 1999;274:16741–16746 [DOI] [PubMed] [Google Scholar]
  • 84. Takaishi H, Konishi H, Matsuzaki H, et al. Regulation of nuclear translocation of forkhead transcription factor AFX by protein kinase B. Proc Natl Acad Sci U S A. 1999;96:11836–11841 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Salih DA, Brunet A. FoxO transcription factors in the maintenance of cellular homeostasis during aging. Curr Opin Cell Biol. 2008;20:126–136 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. van der Horst A, Burgering BM. Stressing the role of FoxO proteins in lifespan and disease. Nat Rev Mol Cell Biol. 2007;8:440–450 [DOI] [PubMed] [Google Scholar]
  • 87. Balaban RS, Nemoto S, Finkel T. Mitochondria, oxidants, and aging. Cell. 2005;120:483–495 [DOI] [PubMed] [Google Scholar]
  • 88. Dröge W. Free radicals in the physiological control of cell function. Physiol Rev. 2002;82:47–95 [DOI] [PubMed] [Google Scholar]
  • 89. Wanagat J, Dai DF, Rabinovitch P. Mitochondrial oxidative stress and mammalian healthspan. Mech Ageing Dev. 2010;131:527–535 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Lean JM, Davies JT, Fuller K, et al. A crucial role for thiol antioxidants in estrogen-deficiency bone loss. J Clin Invest. 2003;112:915–923 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Jagger CJ, Lean JM, Davies JT, Chambers TJ. Tumor necrosis factor-alpha mediates osteopenia caused by depletion of antioxidants. Endocrinology. 2005;146:113–118 [DOI] [PubMed] [Google Scholar]
  • 92. Tyner SD, Venkatachalam S, Choi J, et al. p53 mutant mice that display early ageing-associated phenotypes. Nature. 2002;415:45–53 [DOI] [PubMed] [Google Scholar]
  • 93. de Boer J, Andressoo JO, de Wit J, et al. Premature aging in mice deficient in DNA repair and transcription. Science. 2002;296: 1276–1279 [DOI] [PubMed] [Google Scholar]
  • 94. Nojiri H, Saita Y, Morikawa D, et al. Cytoplasmic superoxide causes bone fragility owing to low-turnover osteoporosis and impaired collagen cross-linking. J Bone Miner Res. 2011;26:2682–2694 [DOI] [PubMed] [Google Scholar]
  • 95. Ambrogini E, Almeida M, Martin-Millan M, et al. FoxO-mediated defense against oxidative stress in osteoblasts is indispensable for skeletal homeostasis in mice. Cell Metab. 2010;11:136–146 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Rached MT, Kode A, Xu L, et al. FoxO1 is a positive regulator of bone formation by favoring protein synthesis and resistance to oxidative stress in osteoblasts. Cell Metab. 2010;11:147–160 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Monroe DG, McGee-Lawrence ME, Oursler MJ, Westendorf JJ. Update on Wnt signaling in bone cell biology and bone disease. Gene. 2012;492:1–18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Clevers H, Nusse R. Wnt/β-catenin signaling and disease. Cell. 2012;149:1192–1205 [DOI] [PubMed] [Google Scholar]
  • 99. Hoogeboom D, Essers MA, Polderman PE, Voets E, Smits LM, Burgering BM. Interaction of FOXO with beta-catenin inhibits beta-catenin/T cell factor activity. J Biol Chem. 2008;283:9224–9230 [DOI] [PubMed] [Google Scholar]
  • 100. Liu H, Fergusson MM, Wu JJ, et al. Wnt signaling regulates hepatic metabolism. Sci Signal. 2011;4:ra6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Iyer S, Ambogini E, Han L, et al. Combined deletion of the transcription factors FoxO1, 3, and 4 from osteoblast progenitors expressing osterix causes bone anabolism and postpones the adverse effects of aging on bone. J Bone Miner Res. 2011;26:1168 (abstract). [Google Scholar]
  • 102. Talchai C, Xuan S, Kitamura T, DePinho RA, Accili D. Generation of functional insulin-producing cells in the gut by Foxo1 ablation. Nat Genet. 2012;44:406–412–S1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Manolagas SC, Almeida M. Gone with the Wnts: beta-catenin, T-cell factor, forkhead box O, and oxidative stress in age-dependent diseases of bone, lipid, and glucose metabolism. Mol Endocrinol. 2007;21:2605–2614 [DOI] [PubMed] [Google Scholar]
  • 104. Hoogeboom D, Burgering BM. Should I stay or should I go: beta-catenin decides under stress. Biochim Biophys Acta. 2009;1796:63–74 [DOI] [PubMed] [Google Scholar]
  • 105. van der Vos KE, Coffer PJ. FOXO-binding partners: it takes two to tango. Oncogene. 2008;27:2289–2299 [DOI] [PubMed] [Google Scholar]
  • 106. DeCarolis NA, Wharton KA, Jr, Eisch AJ. Which way does the Wnt blow? Exploring the duality of canonical Wnt signaling on cellular aging. BioEssays. 2008;30:102–106 [DOI] [PubMed] [Google Scholar]
  • 107. Jin T. The WNT signalling pathway and diabetes mellitus. Diabetologia. 2008;51:1771–1780 [DOI] [PubMed] [Google Scholar]
  • 108. Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell. 2008;132:27–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. He C, Klionsky DJ. Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet. 2009;43:67–93 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Geng J, Klionsky DJ. The Atg8 and Atg12 ubiquitin-like conjugation systems in macroautophagy. ‘Protein modifications: beyond the usual suspects’ review series. EMBO Rep. 2008;9:859–864 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Komatsu M, Waguri S, Ueno T, et al. Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice. J Cell Biol. 2005;169:425–434 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Yen WL, Klionsky DJ. How to live long and prosper: autophagy, mitochondria, and aging. Physiology (Bethesda). 2008;23:248–262 [DOI] [PubMed] [Google Scholar]
  • 113. Vellai T, Takács-Vellai K, Sass M, Klionsky DJ. The regulation of aging: does autophagy underlie longevity? Trends Cell Biol. 2009;19:487–494 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Hansen M, Chandra A, Mitic LL, Onken B, Driscoll M, Kenyon C. A role for autophagy in the extension of lifespan by dietary restriction in C. elegans. PLoS Genet. 2008;4:e24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Wu JJ, Quijano C, Chen E, et al. Mitochondrial dysfunction and oxidative stress mediate the physiological impairment induced by the disruption of autophagy. Aging (Albany NY). 2009;1:425–437 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Salminen A, Kaarniranta K. Regulation of the aging process by autophagy. Trends Mol Med. 2009;15:217–224 [DOI] [PubMed] [Google Scholar]
  • 117. Kenyon CJ. The genetics of ageing. Nature. 2010;464:504–512 [DOI] [PubMed] [Google Scholar]
  • 118. Kume S, Uzu T, Horiike K, et al. Calorie restriction enhances cell adaptation to hypoxia through Sirt1-dependent mitochondrial autophagy in mouse aged kidney. J Clin Invest. 2010;120:1043–1055 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Lipinski MM, Zheng B, Lu T, et al. Genome-wide analysis reveals mechanisms modulating autophagy in normal brain aging and in Alzheimer’s disease. Proc Natl Acad Sci U S A. 2010;107:14164–14169 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Caramés B, Taniguchi N, Otsuki S, Blanco FJ, Lotz M. Autophagy is a protective mechanism in normal cartilage, and its aging-related loss is linked with cell death and osteoarthritis. Arthritis Rheum. 2010;62:791–801 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Simonsen A, Cumming RC, Brech A, Isakson P, Schubert DR, Finley KD. Promoting basal levels of autophagy in the nervous system enhances longevity and oxidant resistance in adult Drosophila. Autophagy. 2008;4:176–184 [DOI] [PubMed] [Google Scholar]
  • 122. Jung HS, Chung KW, Won Kim J, et al. Loss of autophagy diminishes pancreatic beta cell mass and function with resultant hyperglycemia. Cell Metab. 2008;8:318–324 [DOI] [PubMed] [Google Scholar]
  • 123. Ebato C, Uchida T, Arakawa M, et al. Autophagy is important in islet homeostasis and compensatory increase of beta cell mass in response to high-fat diet. Cell Metab. 2008;8:325–332 [DOI] [PubMed] [Google Scholar]
  • 124. Pua HH, Guo J, Komatsu M, He YW. Autophagy is essential for mitochondrial clearance in mature T lymphocytes. J Immunol. 2009;182:4046–4055 [DOI] [PubMed] [Google Scholar]
  • 125. Singh R, Kaushik S, Wang Y, et al. Autophagy regulates lipid metabolism. Nature. 2009;458:1131–1135 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Mortensen M, Ferguson DJ, Edelmann M, et al. Loss of autophagy in erythroid cells leads to defective removal of mitochondria and severe anemia in vivo. Proc Natl Acad Sci USA. 2010;107:832–837 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Masiero E, Agatea L, Mammucari C, et al. Autophagy is required to maintain muscle mass. Cell Metab. 2009;10:507–515 [DOI] [PubMed] [Google Scholar]
  • 128. Dröge W, Schipper HM. Oxidative stress and aberrant signaling in aging and cognitive decline. Aging Cell. 2007;6:361–370 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Onal M, Piemontese M, Xiong J, Wang Y, Han L, Ye S, Komatsu M, Selig M, Weinstein RS, Zhao H, Jilka RL, Almeida M, Manolagas SC, O'Brien CA. Suppression of Autophagy in Osteocytes Mimics Skeletal Aging. J Biol Chem. 2013. 10.1074/ jbc.M112.444190 [DOI] [PMC free article] [PubMed]
  • 130. Onal M, Xiong J, Ye S, et al. Suppression of autophagy in osteoblasts and osteocytes increases oxidative stress and recapitulates the effects of aging on the murine skeleton. J Bone Miner Res. 2012;27:S349 (abstract). [Google Scholar]
  • 131. Mortensen M, Soilleux EJ, Djordjevic G, et al. The autophagy protein Atg7 is essential for hematopoietic stem cell maintenance. J Exp Med. 2011;208:455–467 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Warr MR, Binnewies M, Flach J, et al. FOXO3A directs a protective autophagy program in haematopoietic stem cells. Nature. 2013;494:323–327 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Riggs BL, Khosla S, Melton LJ., III Sex steroids and the construction and conservation of the adult skeleton. Endocr Rev. 2002;23:279–302 [DOI] [PubMed] [Google Scholar]
  • 134. Nakamura T, Imai Y, Matsumoto T, et al. Estrogen prevents bone loss via estrogen receptor alpha and induction of Fas ligand in osteoclasts. Cell. 2007;130:811–823 [DOI] [PubMed] [Google Scholar]
  • 135. Martin-Millan M, Almeida M, Ambrogini E, et al. The estrogen receptor-alpha in osteoclasts mediates the protective effects of estrogens on cancellous but not cortical bone. Mol Endocrinol. 2010;24:323–334 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Almeida M, Iyer S, Martin-Millan M, et al. Estrogen receptor-α signaling in osteoblast progenitors stimulates cortical bone accrual. J Clin Invest. 2013;123:394–404 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Jilka RL. Cytokines, bone remodeling, and estrogen deficiency: a 1998 update. Bone. 1998;23:75–81 [DOI] [PubMed] [Google Scholar]
  • 138. Onal M, Xiong J, Chen X, et al. Receptor activator of nuclear factor κB ligand (RANKL) protein expression by B lymphocytes contributes to ovariectomy-induced bone loss. J Biol Chem. 2012;287: 29851–29860 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Weinstein RS. Glucocorticoid-induced bone disease. N Engl J Med. 2011;365:62–70 [DOI] [PubMed] [Google Scholar]
  • 140. Almeida M, Han L, Ambrogini E, Weinstein RS, Manolagas SC. Glucocorticoids and tumor necrosis factor α increase oxidative stress and suppress Wnt protein signaling in osteoblasts. J Biol Chem. 2011;286:44326–44335 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Plotkin LI, Manolagas SC, Bellido T. Glucocorticoids induce osteocyte apoptosis by blocking focal adhesion kinase-mediated survival. Evidence for inside-out signaling leading to anoikis. J Biol Chem. 2007;282:24120–24130 [DOI] [PubMed] [Google Scholar]
  • 142. Wang FS, Ko JY, Yeh DW, Ke HC, Wu HL. Modulation of Dickkopf-1 attenuates glucocorticoid induction of osteoblast apoptosis, adipocytic differentiation, and bone mass loss. Endocrinology. 2008;149:1793–1801 [DOI] [PubMed] [Google Scholar]
  • 143. Smith E, Frenkel B. Glucocorticoids inhibit the transcriptional activity of LEF/TCF in differentiating osteoblasts in a glycogen synthase kinase-3beta-dependent and -independent manner. J Biol Chem. 2005;280:2388–2394 [DOI] [PubMed] [Google Scholar]
  • 144. Jia D, O’Brien CA, Stewart SA, Manolagas SC, Weinstein RS. Glucocorticoids act directly on osteoclasts to increase their life span and reduce bone density. Endocrinology. 2006;147:5592–5599 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Vedi S, Elkin SL, Compston JE. A histomorphometric study of cortical bone of the iliac crest in patients treated with glucocorticoids. Calcif Tissue Int. 2005;77:79–83 [DOI] [PubMed] [Google Scholar]
  • 146. Cooper MS. 11beta-Hydroxysteroid dehydrogenase: a regulator of glucocorticoid response in osteoporosis. J Endocrinol Invest. 2008;31:16–21 [PubMed] [Google Scholar]
  • 147. Weinstein RS, Wan C, Liu Q, et al. Endogenous glucocorticoids decrease skeletal angiogenesis, vascularity, hydration, and strength in aged mice. Aging Cell. 2010;9:147–161 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Akune T, Ohba S, Kamekura S, et al. PPARgamma insufficiency enhances osteogenesis through osteoblast formation from bone marrow progenitors. J Clin Invest. 2004;113:846–855 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Kang S, Bennett CN, Gerin I, Rapp LA, Hankenson KD, Macdougald OA. Wnt signaling stimulates osteoblastogenesis of mesenchymal precursors by suppressing CCAAT/enhancer-binding protein alpha and peroxisome proliferator-activated receptor gamma. J Biol Chem. 2007;282:14515–14524 [DOI] [PubMed] [Google Scholar]
  • 150. Okamura M, Kudo H, Wakabayashi K, et al. COUP-TFII acts downstream of Wnt/beta-catenin signal to silence PPARgamma gene expression and repress adipogenesis. Proc Natl Acad Sci U S A. 2009;106:5819–5824 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Song L, Liu M, Ono N, Bringhurst FR, Kronenberg HM, Guo J. Loss of wnt/β-catenin signaling causes cell fate shift of preosteoblasts from osteoblasts to adipocytes. J Bone Miner Res. 2012;27:2344–2358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Sharma C, Pradeep A, Wong L, Rana A, Rana B. Peroxisome proliferator-activated receptor gamma activation can regulate beta-catenin levels via a proteasome-mediated and adenomatous polyposis coli-independent pathway. J Biol Chem. 2004;279:35583–35594 [DOI] [PubMed] [Google Scholar]
  • 153. Schneider C, Porter NA, Brash AR. Routes to 4-hydroxynonenal: fundamental issues in the mechanisms of lipid peroxidation. J Biol Chem. 2008;283:15539–15543 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Klein RF, Allard J, Avnur Z, et al. Regulation of bone mass in mice by the lipoxygenase gene Alox15. Science. 2004;303:229–232 [DOI] [PubMed] [Google Scholar]
  • 155. Brodeur MR, Brissette L, Falstrault L, Ouellet P, Moreau R. Influence of oxidized low-density lipoproteins (LDL) on the viability of osteoblastic cells. Free Radic Biol Med. 2008;44:506–517 [DOI] [PubMed] [Google Scholar]
  • 156. Klein BY, Rojansky N, Ben-Yehuda A, Abou-Atta I, Abedat S, Friedman G. Cell death in cultured human Saos2 osteoblasts exposed to low-density lipoprotein. J Cell Biochem. 2003;90:42–58 [DOI] [PubMed] [Google Scholar]
  • 157. Huang MS, Morony S, Lu J, et al. Atherogenic phospholipids attenuate osteogenic signaling by BMP-2 and parathyroid hormone in osteoblasts. J Biol Chem. 2007;282:21237–21243 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Hauschka PV, Mavrakos AE, Iafrati MD, Doleman SE, Klagsbrun M. Growth factors in bone matrix. Isolation of multiple types by affinity chromatography on heparin-sepharose. J Biol Chem. 1986;261:12665–12674 [PubMed] [Google Scholar]
  • 159. Rosen CJ, Donahue LR, Hunter SJ. Insulin-like growth factors and bone: the osteoporosis connection. Proc Soc Exp Biol Med. 1994;206:83–102 [DOI] [PubMed] [Google Scholar]
  • 160. Kawai M, Rosen CJ. The IGF-I regulatory system and its impact on skeletal and energy homeostasis. J Cell Biochem. 2010;111: 14–19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Hayden JM, Mohan S, Baylink DJ. The insulin-like growth factor system and the coupling of formation to resorption. Bone. 1995;17 (2 suppl):93S–98S [DOI] [PubMed] [Google Scholar]
  • 162. Zhang M, Xuan S, Bouxsein ML, et al. Osteoblast-specific knockout of the insulin-like growth factor (IGF) receptor gene reveals an essential role of IGF signaling in bone matrix mineralization. J Biol Chem. 2002;277:44005–44012 [DOI] [PubMed] [Google Scholar]
  • 163. Mohan S, Baylink DJ. Serum insulin-like growth factor binding protein (IGFBP)-4 and IGFBP-5 levels in aging and age-associated diseases. Endocrine. 1997;7:87–91 [DOI] [PubMed] [Google Scholar]
  • 164. Seck T, Scheppach B, Scharla S, et al. Concentration of insulin-like growth factor (IGF)-I and -II in iliac crest bone matrix from pre- and postmenopausal women: relationship to age, menopause, bone turnover, bone volume, and circulating IGFs. J Clin Endocrinol Metab. 1998;83:2331–2337 [DOI] [PubMed] [Google Scholar]
  • 165. Garnero P, Sornay-Rendu E, Delmas PD. Low serum IGF-1 and occurrence of osteoporotic fractures in postmenopausal women. Lancet. 2000;355:898–899 [DOI] [PubMed] [Google Scholar]
  • 166. Jenny NS. Inflammation in aging: cause, effect, or both? Discov Med. 2012;13:451–460 [PubMed] [Google Scholar]
  • 167. Corbi G, Conti V, Scapagnini G, Filippelli A, Ferrara N. Role of sirtuins, calorie restriction and physical activity in aging. Front Biosci (Elite Ed). 2012;4:768–778 [DOI] [PubMed] [Google Scholar]
  • 168. Boyce BF, Schwarz EM, Xing L. Osteoclast precursors: cytokine-stimulated immunomodulators of inflammatory bone disease. Curr Opin Rheumatol. 2006;18:427–432 [DOI] [PubMed] [Google Scholar]

Articles from The Journals of Gerontology Series A: Biological Sciences and Medical Sciences are provided here courtesy of Oxford University Press

RESOURCES