Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Jun 3.
Published in final edited form as: J Bone Miner Res. 2013 Jul;28(7):1509–1518. doi: 10.1002/jbmr.1969

Bone and Skeletal Muscle: Neighbors With Close Ties

Douglas J DiGirolamo 1, Douglas P Kiel 2, Karyn A Esser 3
PMCID: PMC4892934  NIHMSID: NIHMS786363  PMID: 23630111

Abstract

The musculoskeletal system evolved in mammals to perform diverse functions that include locomotion, facilitating breathing, protecting internal organs, and coordinating global energy expenditure. Bone and skeletal muscles involved with locomotion are both derived from somitic mesoderm and accumulate peak tissue mass synchronously, according to genetic information and environmental stimuli. Aging results in the progressive and parallel loss of bone (osteopenia) and skeletal muscle (sarcopenia) with profound consequences for quality of life. Age-associated sarcopenia results in reduced endurance, poor balance, and reduced mobility that predispose elderly individuals to falls, which more frequently result in fracture because of concomitant osteoporosis. Thus, a better understanding of the mechanisms underlying the parallel development and involution of these tissues is critical to developing new and more effective means to combat osteoporosis and sarcopenia in our increasingly aged population. This perspective highlights recent advances in our understanding of mechanisms coupling bone and skeletal muscle mass, and identify critical areas where further work is needed.

Keywords: MUSCLE, BONE, INTERACTION, OSTEOPOROSIS, SARCOPENIA

Introduction

The musculoskeletal system is of paramount importance in our daily lives. In addition to the commonly identified actions of bone and muscle to support upright stance, facilitate movement and breathing, and serve as a protector of our internal organs, these tissues also serve critical metabolic roles. Bone serves as an internal reservoir for calcium to ensure the proper function of nerves and muscle,(1) and skeletal muscle is responsible for over 80% of carbohydrate storage.(2) Recent studies suggest that the skeleton also contributes to glucose homeostasis, further intertwining the actions of bone and muscle beyond locomotion.(3)

Mammalian bone and skeletal muscle involved with locomotion develop in close association from the somitic mesoderm, and accumulate their final adult mass according to specific genetic instructions and environmental cues. Changes in muscle and bone mass brought about by exercise, disuse, or aging are also tightly correlated in both humans and experimental animal models. However, the precise mechanisms responsible for this synchronization remain unclear. It has been widely assumed that coordination of muscle and bone mass occurs through muscle force-generated mechanical signals, which transduce anabolic activity in the adjacent bone.(4) Indeed, evidence from human and animal studies supports the role of mechanical signals as a factor coordinating muscle and bone volume, as is discussed in detail here. The shared mesodermal origin of muscle and bone(5) presents an additional possibility; that common molecular networks serve to coordinate their mass.(611)

Clearly, a better understanding of how muscle and bone synchronize their mass throughout life is critical to the development of more effective strategies to improve musculo-skeletal health and function. In this regard, many of the studies we review herein suggest a dominant role of muscle over bone in synchronizing the mass of these two tissues (at least in postnatal life). Such a hierarchy of control in synchronizing the musculo-skeletal system, if brought to bear through additional study, could completely shift our paradigm for treating osteoporosis and frailty. In this perspective, we review recent findings regarding the mechanisms that coordinate the mass of muscle and bone—from early development through aging and involution—and discuss how these data might help guide our approaches to treating disorders in both tissues.

Developmental Biology of the Musculoskeletal System

Locomotion in vertebrates requires muscles to contract and work against the levers of an internal skeleton, enabling the movement of body parts. The vertebrate skeleton is composed of bone and cartilage linked to skeletal muscle through tendons (for an excellent review of tendons in musculoskeletal development see Schweitzer and colleagues(12)). The skeleton forms in a discrete stepwise process initiated by condensation of mesodermal mesenchymal precursors at the future sites of bone. Following condensation, these precursors differentiate into chondrocytes to form a cartilage anlage (endochondral bone formation) or directly into osteoblasts to form bone (intramembranous bone formation), depending upon positional cues.(13) Once formed, the skeleton is continually remodeled throughout life, which allows for repair of microdamage and adaptive response to increased or decreased mechanical loads. The process of bone remodeling is achieved by the coordinated actions of the bone-forming osteoblasts, bone-resorbing osteoclasts, and osteocytes—terminally differentiated cells of the osteoblast lineage that are embedded in mineralized bone and appear to serve a multitude of functions including mechano-sensation, regulating bone remodeling (via receptor activator of NF-kB ligand [RANKL] and Sclerostin), and participating in phosphate homeostasis (via fibroblast growth factor 23 [FGF23]).(14) The rate and degree of coupling of bone formation to resorption during remodeling is regulated by autocrine, paracrine, and endocrine factors including, but not limited to, Wnts, Hedgehog, and Notch, bone morphogenetic protein (BMP) families, transforming growth factor-β (TGF-β), growth hormone (GH), and insulin-like growth factor-1 (IGF-1), FGF-2, interleukin-6 (IL-6) type cytokines, and ephrinB2 and B4.(15)

Myogenesis occurs immediately adjacent to, and concurrent with, the development of the skeleton during embryogenesis. In the case of skeletal muscle in the trunk and limbs, precursors from the paraxial mesoderm differentiate and fuse to form multinucleated syncytia, or myofibers, that comprise skeletal muscle.(16) Specification of mesodermal precursor cells into the myogenic lineage occurs in the somite for limb muscles, under the control of Pax3/7.(17) Signals from surrounding tissues then increase myoblast expression of myogenic factors (Myf5/MyoD) and drive further differentiation and myogenesis.(17,18) During primary myogenesis, myoblasts fuse to form nascent myotubes, with relatively few nuclei. Secondary myogenesis is characterized by subsequent recruitment and fusion of additional myoblasts and ultimately gives rise to the multinucleated, mature myofibers. However, it would appear that not all Myf5/MyoD expressing myoblasts fuse. Muscle satellite cells (which are Pax7-positive and recently demonstrated to have previously expressed Myf5 and MyoD) can first be observed under the basal lamina of muscle fibers in late fetal stages. These satellite cells do not divide and remain on the periphery of the myofibers to serve as the source of new myonuclei during postnatal growth and injury repair.(17,18) Similar to the regulation of bone development and mass, skeletal muscle development and maintenance is regulated by morphogens and growth factors, many of which overlap with those involved in skeletogenesis, such as Wnts, Hedgehog, Notch, FGFs, IGF-1, and TGF-β.(16) Among the most dominant factors controlling muscle size is myostatin, a member of the TGF-b superfamily. Myostatin negatively regulates skeletal muscle size by activating ACVR2B and initiating Smad2/3 signaling.(19,20)

Many of the overlapping signaling pathways present in muscle and bone development exert similar functions in both tissues. For example, GH and IGF-1 increase proliferation and differentiation of both osteoblasts(21) and myoblasts(22) during development in mice. Some differences are apparent, however, in the activity of these growth factors between muscle and bone. GH appears to uniquely interact with sex steroids in determining the sexually dimorphic patterns of the skeleton.(23)

Wnt signaling is also critical for the development of both muscle and bone and exhibits significant overlap in function between the two tissues. In muscle, Wnts control myogenic regulatory factor (MRF) expression during early embryogenesis to initiate the myogenic program, as well as regulating satellite cell differentiation, self renewal, and muscle fiber growth in response to loading (the latter two through noncanonical signaling) in adult life.(24) Similarly in bone, Wnt signaling is critical for the specification of mesenchymal progenitors toward the osteoblast lineage and responding to mechanical loading.(25) Interestingly, the precise control of Wnts in regulating development seems to be dependent upon timing of exposure in bone,(25) whereas in muscle, the level of Wnt signaling seems to be the primary modulator.(24) A better understanding of the molecular bases underlying the divergent roles in these common signaling pathways of muscle and bone might reveal novel therapeutic targets.

The close physical linkage of bone and muscle development is evident even before birth. For example, the varying circumferential shape of different long bones occurs through asymmetric mineral deposition, apparently in response to site-specific mechanical strains applied to the newly forming bones by their associated muscle groups in utero.(26) In agreement with this notion, the shapes of long bones from muscular dysgenesis (mdg) mice, which lack muscular contraction due to an excitation-contraction coupling defect, are nearly uniformly circular and mechanically inferior to those of normal mice. Muscular contraction during embryogenesis has also been demonstrated to be critical for maintaining specification of joint progenitors to ensure proper joint morphogenesis. A study from Kahn and colleagues(27) noted the loss of elbow, midcarpal, and hip joints in three different mutant mice that lacked muscle, as well as the mdg mice described in the study by Sharir et al.(26) Evidence supporting the importance of muscular contraction for proper skeletal development can also be observed in rare neuromuscular disorders that reduce human fetal muscle contraction. Fetal immobilization secondary to congenital myotonic dystrophy or spinal muscular atrophy resulted in thin, hypomineralized, and elongated long bones with multiple fractures.(28,29) In addition to mechanical crosstalk during embryogenesis and fetal development, recent evidence suggests bone and muscle development are intimately linked through morphogen signaling. In both mouse and chick embryos, bone-derived Indian hedgehog (Ihh) promotes fetal myoblast survival and secondary myogenesis.(30) Importantly, the ability of bone-derived Ihh to support myogenesis indicates that muscle-bone crosstalk is bidirectional.

Postnatal Coordination of Bone and Muscle Mass

During postnatal growth in mammals, bone and muscle mass increase dramatically and proportionally, achieving peak mass at around the same time (25–35 years old in humans).(31) Longitudinal growth of long bones occurs through endochondral ossification.(32) The growth plate generates a cartilaginous template that becomes new trabecular bone, elongating the metaphysis. Trabeculae near the outer edges of the bone eventually coalesce to form the metaphyseal cortex. As the bone elongates, those trabeculae near the center of long bones are resorbed to form the marrow cavity. In the diaphysis, cross-sectional growth is mediated by the combination of periosteal cortical apposition and endosteal resorption. The postnatal growth of muscle results entirely from an increase in muscle fiber size (hypertrophy), although the mechanism(s) driving this process has been widely disputed. Muscle hypertrophy had long been thought to require the proliferative activity of satellite cells and their fusion with existing muscle fibers,(3335) but recent evidence has demonstrated hypertrophy of muscle fibers in the absence of satellite cells.(36)

Genetic background

A fundamental determinant of peak bone and muscle mass is genetic background. Bivariate linkage analysis in large human populations has identified significant quantitative trait loci (QTLs) shared by leg lean mass with shaft cross-sectional area on chromosome 12p12–12p13 and with neck shaft angle on 14q21–22.(37) Another study of 102 monozygotic and 113 dizygotic older female twin pairs demonstrated shared genetic components in muscle cross-sectional area of the lower leg, bending strength of the tibial shaft, and compressive strength of the distal tibia from pQCT scans.(38) Analogous studies in animal models also indicate that common gene subsets control bone and muscle mass. For example, inactivating mutations of myostatin cause hypermuscularity in mice, with increased cortical bone mineral content (BMC) at the L5 vertebra, larger spinous processes, and larger entheses on the femur and humerus.(39,40) It has also been proposed that genetic background might determine responsivity of the muscle-bone unit to mechanical stimuli. In support of this idea, Lang and colleagues(41) used structural equation modeling to examine the extent to which select genetic loci manifest their pleiotropic effects in the musculoskeletal system through adaptation to mechanical stimuli. Genetic analysis in male and female F2 offspring from a B6XD2 intercross demonstrated correlations among bone strength, muscle mass, and physical activity, and identified several QTLs associated with mechanosensitivity.

Sex steroids

Superimposed on these genetic determinants of bone and muscle mass are anabolic stimuli that occur postnatally, the most dominant of which is puberty. During the pubertal growth spurt, bone and muscle mass accumulate rapidly under the influence GH, IGF-1, and sex hormones.(42) Pubertal increases in lean body mass are detected prior to increased BMC, suggesting that skeletal mass increases to accommodate stresses incurred from increased muscle force.(43) In agreement with this concept, tibial cross-sectional moment of inertia is tightly correlated with the cross-sectional area of the calf muscles.(44) Although females enter puberty first, males have a longer pubertal growth spurt and greater peak longitudinal growth velocity than females, which ultimately results in 10% greater height and 25% greater peak bone mass in males.(45) In addition, male bones attain a larger diameter due to greater periosteal expansion and less endocortical apposition than females.(46,47) These sex-specific differences in skeletal acquisition are mediated by the differential effects of androgens and estrogen.(48) Peak muscle mass is also higher in males(49,50) as a result of the well documented anabolic effects of androgens in muscle hypertrophy (in both sexes) commonly observed in professional bodybuilding. Additionally, testosterone increases muscle mass and strength in hypogonadal men(51) and normalizes the reduced muscle mass in orchidectomized rodents.(52) By contrast, estrogen appears to have little direct effect on muscle hypertrophy, and replacement therapy fails to prevent loss of muscle mass and strength in aging females.(53,54)

Mechanical forces

In addition to the genetic determinants and humoral factors affecting musculoskeletal mass, and likely in direct interplay with them, the level of physical activity in which a human or animal engages plays a tremendous role in determining postnatal muscle and bone mass. Physical activity exerts anabolic effects on the skeleton either directly, indirectly through mechanical forces generated by muscle action, or indirectly through endocrine regulation (ie, elevation of GH and IGF-1), and understanding the precise mechanisms underlying this anabolic response is an area of very active research. Exaggerated examples of mechanical effects achieved though vigorous exercise are seen in the increased bone density of the dominant arm (which also has increased muscle mass) of premier racquet sport participants.(5557) Thankfully for the majority, it appears that such benefits are not restricted to elite athletes. Both muscle and bone have recently been demonstrated to be responsive to low-magnitude mechanical signals.(58) Genetic linkage studies in mice have been performed to tease out genes that may be responsible for the anabolic response of bone to mechanical stimulation. For example, Kesavan and colleagues(59) performed QTL analysis after applying bending loads to the tibias of 10 week-old female F2 mice from a B6XC3H intercross and identified several loci that appeared responsible for mechano-sensitivity. Whether these same genes exert such effects in muscle is a question that clearly warrants further investigation.

Conversely, disuse or unweighting of the muscle-bone unit in immobilized individuals (bed rest) or after space flight, respectively, results in a dramatic loss of bone and muscle mass.(60) In many cases, the loss of muscle appears to drive the loss of bone. For example, individuals with Duchenne muscular dystrophy and cerebral palsy—primary defects in muscle function—also have reduced bone mass and increased fracture risk.(6164) Furthermore, significant bone loss occurs in patients with spinal cord injury (SCI)(65); where rapid and profound muscle loss of muscle mass secondary to motor neuron loss is presumably the precipitating factor.(66) Additional evidence supporting a primary role for muscle as a determinant of bone mass comes from studies in subjects exposed to chronic unloading during space flight. Individuals exposed to zero gravity experience minimal neuromuscular mechanical stimulation (bodies in space have mass but not weight), with significant muscular atrophy and bone loss (1% of total muscle and 1.8% to 2% of total bone lost per month(60)). Interestingly, the losses of muscle and bone during space flight are far less pronounced in the upper extremities that do not typically bear weight, but myriad physiologic changes in space (cardiovascular, etc.) make interpretation of such findings difficult.

Other studies in microgravity, or in settings designed to minimize the effects of gravity, suggest that the variable effects of loading and unloading on different regions of the musculo-skeletal system may actually stem from its biomechanical evolution to protect joints against the force of gravity.(67) In this regard, the musculature of the human body can be largely anatomically divided into monoarticulate (muscles that cross a single joint) or biarticulate (muscles that cross two joints). In a complex movement, such as pedaling a bicycle, monoarticulate muscles generally maintain joint position while biarticulate muscles orchestrate the direction and force of leg movement.(68) Using a ballistic knee movement model, Richardson and Bullock(69) experimentally eliminated gravitational load cues from subjects’ knee movements and demonstrated that biarticulate muscle recruitment increased with increasing speed, while monoarticulate muscles were unaffected. Conversely, activation of monoarticulate muscles in the knee and hip was significantly higher under weight bearing versus non–weight bearing conditions, with normal gravitational load.(70) This suggested the presence of an “antigravity” system within the musculature, particularly monoarticulate, to support and protect joints and maintain posture under gravitational load. Indeed, in conditions of microgravity, such postural muscles demonstrate the greatest losses of mass.(71,72) The loss of muscle mass from these postural muscles in unloaded conditions is also associated with fiber-type switching—from slow (endurance) to fast (easily fatigued) fibers.(73) Similar changes are also observed in cases of immobility and bed rest.(74) It should be noted that age-related fiber-type switching differs from that seen in microgravity or during bed rest; ie, the muscles that atrophy tend to be type IIX (fast) fibers with increasing dependence on type IIA (intermediate) and type I (slow) fibers.(75) Thus, it is reasonable to suggest that aging and bed rest simultaneously impinge upon two different fiber types, compounding the impairment of muscle function. The atrophy of postural and stabilizing muscles during a hospitalization (and switch to easily fatigued fast fibers) might predispose an elderly individual to poor(er) posture and biomechanics, which could lead to joint damage or even falls (because the age-related switch from fast fibers would make it more difficult to compensate for a loss of balance), creating a vicious cycle of muscle and bone loss from further inactivity.

Experimental maneuvers to mimic unloading in rodents, such as tail suspension, can reproduce some of the effects of microgravity, including rapid loss of muscle and bone.(76) Interestingly, certain genetic strains of mice (C3H/HeJ) are resistant to unloading-induced bone loss.(77) These models are beginning to be used to examine the underlying genetic determinants of responsiveness of muscle and bone to unweighting, with obvious implications for identifying potential therapeutic targets to prevent muscle and bone loss associated with more “down to earth” problems like injury and immobility.

Aging and Musculoskeletal Involution

The mass of both skeletal muscle and bone are profoundly affected by age. Age-related muscle atrophy, referred to as “sarcopenia,” is characterized by the loss of both strength and skeletal muscle mass.(78) Muscle mass decreases by 3% to 8% per decade after age 30 years, and the pace of muscle loss only quickens after age 60 years.(79) This loss of muscle mass and strength is due to progressive atrophy, loss of muscle fibers, reduced motor neuron input, and impaired function of the contractile apparatus within each fiber.(80) Aging is further complicated by periods of bed rest or inactivity, due to an injury such as a hip fracture, which can result in profound losses of both bone and muscle in parallel.(81) In a sample of community-dwelling seniors, hospitalizations were associated with a loss of lean mass and fat mass, as well as a loss of strength in men.(82) Even in healthy older persons, 10 days of bed rest can produce losses of strength of over 13% along with losses of aerobic capacity.(83) Bed rest might also have deleterious effects on postural support that could predispose to joint damage and/or falls.(67) Although the declines in muscle can be partially explained by reduced physical activity with age, sarcopenia also involves metabolic abnormalities, including reduced insulin sensitivity, fat and connective tissue infiltration, impaired oxidative defense, reduced hormone levels, and decreased mitochondrial activity(8486) that further confound muscle function.

At the cellular level, these sequelae of sarcopenia can disturb the already faltering balance of protein synthesis and degradation present in aging muscle. Muscle atrophy occurs through the concerted actions of numerous signaling pathways and molecular mechanisms (recently reviewed by Bonaldo and Sandri(87)) including insulin-like growth factor 1 (IGF-1)-Akt- mammalian target of rapamycin (mTOR)/Forkhead box O (FoxO), inflammatory cytokines and NF-kB signaling, myostatin/activin signaling, the ubiquitin-proteasome system, and the autophagy- lysosome system. Indeed, recent studies suggest that autophagy may play a significant role in sarcopenic muscle atrophy. Autophagy is critical in many cell types for the turnover of cellular components, both on a continual basis and in response to stress, nutrient deprivation, or cytokines.(88) Evidence of muscle diseases in states of both excess(89) and defective autophagy(90) suggest that there may be an optimum autophagy flux in muscle to maintain contractile function, and disruption of normal autophagy with age might predispose muscle fibers to chronic contractile damage and, eventually, atrophy.(91) As noted above, mitochondrial function declines with age and results in progressive activation of autophagy to recycle these dysfunctional mitochondria, possibly causing just such a disruption in autophagy flux. In support of this notion, transgenic expression of peroxisome proliferator-activated receptor gamma coactivator-1a (PGC1a) in skeletal muscle (a master regulator of mitochondrial biogenesis) prevented the age-related increase in autophagy and loss of muscle mass in mice.(92)

In addition to these changes within the myofiber, muscle regenerative capacity is further hampered with age due to a reduction in—and impaired function of persisting—satellite cells.(93) As described earlier, quiescent satellite cells reside between the sarcolemma and basal lamina of myofibers and are critical for postnatal growth and regeneration following injury. Although the precise mechanism leading to their decline and dysfunction remains to be proved, evidence from other tissue- resident stem cell populations, including hematopoietic and neural stem cells, implicate chronic exposure to inflammatory factors and oxidative stress as likely factors underlying the disruption of the balance of stem cell proliferation, self-renewal, and appropriate differentiation.(94,95) Interestingly, despite the many indications of intrinsic defects in both myofibers and satellite cells with age, early work by Carlson and Faulkner(96) clearly demonstrated that the aged environment exerts a dominant effect on the regenerative capacity of muscle. In their study, the extensor digitorum longus (EDL) from young rats was transplanted into young or old rats, and vice versa. Surprisingly, the EDL from both young and old rats was able to regenerate to a similar extent in young recipients, whereas even young muscle regenerated poorly in the old recipients.(96) More recently, elegant experiments involving parabioses between young and old mice demonstrated a rejuvenation of satellite cell populations in aged mice when exposed to the youthful shared circulation.(97) These effects of the aged environment on muscle regenerative capacity have been shown to involve a decline in Notch signaling(98) and an increase in circulating Wnt molecules.(99)

Concomitant with the loss of muscle, aging also results in progressive bone loss, leading to bone fragility and increased risk for osteoporosis and fractures. In fact, age-related muscle wasting may coexist with osteoporosis, establishing a vicious cycle between dysfunctional muscle and bone. This age-related loss of bone results from a decreased capacity to effectively remodel itself. Osteoblast numbers and function decline with age(100) in association with decreased levels of sex steroids, GH, and IGF-1.(101103) Interestingly, many of the same mechanisms that appear to impinge upon satellite cells with age also negatively affect the mesenchymal stem cells (MSCs) that give rise to osteoblasts, including oxidative stress.(104) Further, exposing MSCs from aged mice to a decellularized extracellular matrix produced by the MSCs of young mice corrected the proliferative and osteogenic differentiation defects in aged MSCs.(105) Unlike osteoblasts, osteoclast-mediated resorption remains constant, or even increases (by as much as 90% in postmenopausal women).(106) Ineffective remodeling of bone with advanced age is also partially explained by a reduction in responsiveness to mechanical loads,(107) possibly via alterations in mechanosensation by osteocytes.(14) Interestingly, this age-related decline in bone remodeling in response to mechanical load parallels that of the decreased muscle input, highlighting again, the possibility of a dominant role of muscle in the muscle-bone unit.

Body composition changes with aging also include the accumulation of fat viscerally, in the bone marrow, and infiltrating between and within muscle fibers.(108) The entity referred to as “sarcopenic obesity” is the co-occurrence of muscle loss with increases in adiposity, and it appears to be associated with functional disability.(109) There may be an underlying link between sarcopenic obesity and osteoporosis, because both muscle and bone exhibit increased numbers of adipocytes with age. Intramuscular fat has been shown be associated with inflammatory markers,(110) which may represent a mechanism explaining the recent observation that periaortic fat is associated with lower volumetric bone density of the adjacent spine.(111) Thus, the age-related decline in muscle mass and bone mass may be partly linked to adipocyte physiology and their proinflammatory milieu.

Emerging Endocrine Roles

A number of studies support the notion of muscle functioning in an endocrine fashion to affect other tissues and organs, including liver, pancreas, vasculature, fat, and importantly for our discussion, bone (reviewed by Pedersen and Febbraio(112)). These muscle-secreted endocrine factors, termed myokines, include myostatin, leukemia inhibitory factor (LIF), IL-6, IL-7, brain-derived neurotrophic factor (BDNF), IGF-1, FGF-2, follistatin-like protein 1 (FSTL-1), and irisin.(112) It is reasonable to postulate that many myokines could exert direct effects on adjacent—or even distant—bone, especially IGF-1 and FGF-2, given the primary importance of these factors in bone development.(15) Moreover, myokines could also indirectly impact bone through actions on other tissues. For example, IL- 6 can increase the secretion of insulin from the pancreas by increasing GLP-1 secretion from L cells and alpha cells.(113) Insulin could then feed into the recently described bone–pancreas endocrine loop and exert secondary effects upon bone.(3) Furthermore, the ability of muscle secreted irisin to induce brown-fat-like development of white fat(114) could also indirectly impact bone, given the crosstalk between bone and fat.(115,116) Additionally, the recent identification of a novel muscle secretory factor, musclin,(117) which is identical in protein sequence to bone-derived osteocrin,(118) suggests the possibility of an endocrine loop directly connecting muscle and bone.

Summary and Perspectives

Bone and muscle, from early embryonic development through aging and involution, are tightly coupled in both form and function. Numerous factors impact the relative mass of these two tissues, including genetic background, morphogens, sex steroids, other circulating factors (eg, GH and IGF-1), and mechanical forces. Although morphogens and genetic factors may primarily control muscle and bone development during embryogenesis, and sex steroids exert a dominant role during pubertal growth, it appears that mechanical forces exert an overarching control throughout life. In this regard, much of the data reviewed in this Perspective suggests that muscle predominates over bone in synchronizing tissue mass of the musculoskeletal system. That is not to say that communication between muscle and bone is necessarily unidirectional. As described in our discussion of the developmental biology of the musculoskeletal system, bone-derived Ihh is required for normal muscle development in mouse and chick embryogenesis. However, the postnatal setting clearly favors a strong influence of muscle on bone mass through mechanical signals, which likely employ the actions of hormones and morphogens as determined by one's genetic background. Therapeutic approaches to treat age-related and disease-related musculoskeletal deficits based upon this “muscle-predominant” concept are an area of extremely active research, with many focused on targeting the myostatin/activin signaling pathway.(119128) More recent data suggests that components of this pathway may also function directly in osteoblasts in a manner analogous to that of skeletal muscle,(129) and may represent a common molecular means by which bone and muscle coordinate their mass.

In the face of the wealth of evidence supporting a biomechanical link that coordinates bone and muscle mass, many important questions regarding the nature of this putative mechanism remain open. Are the responses to mechanical loading of bone and muscle occurring in parallel, or is muscle the primary responder to physical activity, transducing that signal to bone in a secondary fashion? What is the nature of the mechanical signal posited by the mechanostat theory?(130) Further, if osteocytes serve as the primary mechanosensor, how do they translate mechanical signals into the biochemical orchestration of bone remodeling? Does muscle function as a mechanical-endocrine converter, where the mechanical events experienced by muscle induce paracrine/endocrine effectors, which then influence bone development? This seems an ever more attractive hypothesis given evidence from the rapidly expanding myokine field and our growing understanding of organ system crosstalk in other physiologic contexts. Finally, many of the examples of postnatal coupling of bone and muscle cited in this Perspective are likely to involve some neurological input, given the well-described roles of the nervous system in regulating both normal muscle(61,62,131,132) and bone mass,(133135) and may present an opportunity for unique therapeutic interventions. Answers to the questions above are critical for understanding the coordinate regulation of muscle and bone mass, and for identifying new targets to combat their inevitable decline with age.

Acknowledgments

Authors’ roles: DJD designed and drafted the manuscript. All authors reviewed and contributed to subsequent drafts and approved the final version.

Footnotes

Disclosures

All authors state that they have no conflicts of interest.

References

  • 1.Civitelli R, Ziambaras K. Calcium and phosphate homeostasis: concerted interplay of new regulators. J Endocrinol Invest. 2011;34(7 Suppl):3–7. [PubMed] [Google Scholar]
  • 2.DeFronzo RA, Gunnarsson R, Bjorkman O, Olsson M, Wahren J. Effects of insulin on peripheral and splanchnic glucose metabolism in noninsulin-dependent (type II) diabetes mellitus. J Clin Invest. 1985;76(1):149–55. doi: 10.1172/JCI111938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.DiGirolamo DJ, Clemens TL, Kousteni S. The skeleton as an endocrine organ. Nat Rev Rheumatol. 2012;8(11):674–83. doi: 10.1038/nrrheum.2012.157. [DOI] [PubMed] [Google Scholar]
  • 4.Frost HMA. 2003 update of bone physiology and Wolff's Law for clinicians. Angle Orthod. 2004;74(1):3–15. doi: 10.1043/0003-3219(2004)074<0003:AUOBPA>2.0.CO;2. [DOI] [PubMed] [Google Scholar]
  • 5.Schoenwolf GC, Bleyl SB, Brauer PR, Francis-West PH, editors. Larsen's Human Embryology. 4th edition Churchill Livingstone/Elsevier; Philadelphia: 2009. Chapter 8: Development of the Musculoskeletal System. [Google Scholar]
  • 6.Kaji H. Linkage between muscle and bone: common catabolic signals resulting in osteoporosis and sarcopenia. Curr Opin Clin Nutr Metab Care. 2013 May;16(3):272–7. doi: 10.1097/MCO.0b013e32835fe6a5. [DOI] [PubMed] [Google Scholar]
  • 7.Warden SJ, Galley MR, Richard JS, George LA, Dirks RC, Guildenbecher EA, Judd AM, Robling AG, Fuchs RK. Reduced gravitational loading does not account for the skeletal effect of botulinum toxin-induced muscle inhibition suggesting a direct effect of muscle on bone. Bone. 2013;54(1):98–105. doi: 10.1016/j.bone.2013.01.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Ruschke K, Hiepen C, Becker J, Knaus P. BMPs are mediators in tissue crosstalk of the regenerating musculoskeletal system. Cell Tissue Res. 2012;347(3):521–44. doi: 10.1007/s00441-011-1283-6. [DOI] [PubMed] [Google Scholar]
  • 9.Jahn K, Lara-Castillo N, Brotto L, Mo CL, Johnson ML, Brotto M, Bonewald LF. Skeletal muscle secreted factors prevent glucocorticoid-induced osteocyte apoptosis through activation of beta-catenin. Eur Cell Mater. 2012;24:197–209. doi: 10.22203/ecm.v024a14. discussion 209-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Willett NJ, Li MT, Uhrig BA, Boerckel JD, Huebsch N, Lundgren TL, Warren GL, Guldberg RE. Attenuated human bone morphogenetic protein-2-mediated bone regeneration in a rat model of composite bone and muscle injury. Tissue Eng Part C Methods. 2013;19(4):316–25. doi: 10.1089/ten.tec.2012.0290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Mo C, Romero-Suarez S, Bonewald L, Johnson M, Brotto M. Prostaglandin e2: from clinical applications to its potential role in bone- muscle crosstalk and myogenic differentiation. Recent Pat Biotechnol. 2012;6(3):223–9. doi: 10.2174/1872208311206030223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Schweitzer R, Zelzer E, Volk T. Connecting muscles to tendons: tendons and musculoskeletal development in flies and vertebrates. Development. 2010;137(17):2807–17. doi: 10.1242/dev.047498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Olsen BR, Reginato AM, Wang W. Bone development. Annu Rev Cell Dev Biol. 2000;16:191–220. doi: 10.1146/annurev.cellbio.16.1.191. [DOI] [PubMed] [Google Scholar]
  • 14.Bonewald LF. The amazing osteocyte. J Bone Miner Res. 2011;26(2):229–38. doi: 10.1002/jbmr.320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Kular J, Tickner J, Chim SM, Xu J. An overview of the regulation of bone remodelling at the cellular level. Clin Biochem. 2012 Aug;45(12):863–73. doi: 10.1016/j.clinbiochem.2012.03.021. [DOI] [PubMed] [Google Scholar]
  • 16.Braun T, Gautel M. Transcriptional mechanisms regulating skeletal muscle differentiation, growth and homeostasis. Nat Rev Mol Cell Biol. 2011;12(6):349–61. doi: 10.1038/nrm3118. [DOI] [PubMed] [Google Scholar]
  • 17.Buckingham M, Vincent SD. Distinct and dynamic myogenic populations in the vertebrate embryo. Curr Opin Genet Dev. 2009;19(5):444–53. doi: 10.1016/j.gde.2009.08.001. [DOI] [PubMed] [Google Scholar]
  • 18.Bismuth K, Relaix F. Genetic regulation of skeletal muscle development. Exp Cell Res. 2010;316(18):3081–6. doi: 10.1016/j.yexcr.2010.08.018. [DOI] [PubMed] [Google Scholar]
  • 19.Otto A, Patel K. Signalling and the control of skeletal muscle size. Exp Cell Res. 2010;316(18):3059–66. doi: 10.1016/j.yexcr.2010.04.009. [DOI] [PubMed] [Google Scholar]
  • 20.McPherron AC, Lawler AM, Lee SJ. Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member. Nature. 1997;387(6628):83–90. doi: 10.1038/387083a0. [DOI] [PubMed] [Google Scholar]
  • 21.DiGirolamo DJ, Mukherjee A, Fulzele K, Gan Y, Cao X, Frank SJ, Clemens TL. Mode of growth hormone action in osteoblasts. J Biol Chem. 2007;282(43):31666–74. doi: 10.1074/jbc.M705219200. [DOI] [PubMed] [Google Scholar]
  • 22.Mavalli MD, DiGirolamo DJ, Fan Y, Riddle RC, Campbell KS, van Groen T, Frank SJ, Sperling MA, Esser KA, Bamman MM, Clemens TL. Distinct growth hormone receptor signaling modes regulate skeletal muscle development and insulin sensitivity in mice. J Clin Invest. 2010;120(11):4007–20. doi: 10.1172/JCI42447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Singhal V, Goh BC, Bouxsein ML, Faugere MC, DiGirolamo DJ. Osteoblast-restricted disruption of the growth hormone receptor in mice results in sexually dimorphic skeletal phenotypes. [2013 Apr 22];Bone Res [Internet] 2013 Mar;1(1):85–97. doi: 10.4248/BR201301006. Available from: http://www.boneresearch.org/index.php?c=content&a=show&id=73&fulltext=1. DOI: 10.4248/BR201301006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.von Maltzahn J, Chang NC, Bentzinger CF, Rudnicki MA. Wnt signaling in myogenesis. Trends Cell Biol. 2012;22(11):602–9. doi: 10.1016/j.tcb.2012.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Regard JB, Zhong Z, Williams BO, Yang Y. Wnt signaling in bone development and disease: making stronger bone with Wnts. Cold Spring Harb Perspect Biol. 2012;4(12):a007997. doi: 10.1101/cshperspect.a007997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Sharir A, Stern T, Rot C, Shahar R, Zelzer E. Muscle force regulates bone shaping for optimal load-bearing capacity during embryogenesis. Development. 2011;138(15):3247–59. doi: 10.1242/dev.063768. [DOI] [PubMed] [Google Scholar]
  • 27.Kahn J, Shwartz Y, Blitz E, Krief S, Sharir A, Breitel DA, Rattenbach R, Relaix F, Maire P, Rountree RB, Kingsley DM, Zelzer E. Muscle contraction is necessary to maintain joint progenitor cell fate. Dev Cell. 2009;16(5):734–43. doi: 10.1016/j.devcel.2009.04.013. [DOI] [PubMed] [Google Scholar]
  • 28.Rodriguez JI, Palacios J, Garcia-Alix A, Pastor I, Paniagua R. Effects of immobilization on fetal bone development. A morphometric study in newborns with congenital neuromuscular diseases with intrauterine onset. Calcif Tissue Int. 1988;43(6):335–9. doi: 10.1007/BF02553275. [DOI] [PubMed] [Google Scholar]
  • 29.Rodriguez JI, Garcia-Alix A, Palacios J, Paniagua R. Changes in the long bones due to fetal immobility caused by neuromuscular disease. A radiographic and histological study. J Bone Joint Surg Am. 1988;70(7):1052–60. [PubMed] [Google Scholar]
  • 30.Bren-Mattison Y, Hausburg M, Olwin BB. Growth of limb muscle is dependent on skeletal-derived Indian hedgehog. Dev Biol. 2011;356(2):486–95. doi: 10.1016/j.ydbio.2011.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Bonjour JP, Theintz G, Law F, Slosman D, Rizzoli R. Peak bone mass. Osteoporos Int. 1994;4(Suppl 1):7–13. doi: 10.1007/BF01623429. [DOI] [PubMed] [Google Scholar]
  • 32.Kronenberg HM. Developmental regulation of the growth plate. Nature. 2003;423(6937):332–6. doi: 10.1038/nature01657. [DOI] [PubMed] [Google Scholar]
  • 33.Bruusgaard JC, Johansen IB, Egner IM, Rana ZA, Gundersen K. Myonuclei acquired by overload exercise precede hypertrophy and are not lost on detraining. Proc Natl Acad Sci U S A. 2010;107(34):15111–6. doi: 10.1073/pnas.0913935107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Schiaffino S, Bormioli SP, Aloisi M. The fate of newly formed satellite cells during compensatory muscle hypertrophy. Virchows Arch B Cell Pathol. 1976;21(2):113–8. doi: 10.1007/BF02899148. [DOI] [PubMed] [Google Scholar]
  • 35.Moss FP, Leblond CP. Nature of dividing nuclei in skeletal muscle of growing rats. J Cell Biol. 1970;44(2):459–62. doi: 10.1083/jcb.44.2.459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.McCarthy JJ, Mula J, Miyazaki M, Erfani R, Garrison K, Farooqui AB, Srikuea R, Lawson BA, Grimes B, Keller C, Van Zant G, Campbell KS, Esser KA, Dupont-Versteegden EE, Peterson CA. Effective fiber hypertrophy in satellite cell-depleted skeletal muscle. Development. 2011;138(17):3657–66. doi: 10.1242/dev.068858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Karasik D, Zhou Y, Cupples LA, Hannan MT, Kiel DP, Demissie S. Bivariate genome-wide linkage analysis of femoral bone traits and leg lean mass: Framingham study. J Bone Miner Res. 2009;24(4):710–8. doi: 10.1359/JBMR.081222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Mikkola TM, Sipila S, Rantanen T, Sievanen H, Suominen H, Tiainen K, Kaprio J, Koskenvuo M, Kauppinen M, Heinonen A. Muscle cross-sectional area and structural bone strength share genetic and environmental effects in older women. J Bone Miner Res. 2009;24(2):338–45. doi: 10.1359/jbmr.081008. [DOI] [PubMed] [Google Scholar]
  • 39.Hamrick MW. Increased bone mineral density in the femora of GDF8 knockout mice. Anat Rec A Discov Mol Cell Evol Biol. 2003;272(1):388–91. doi: 10.1002/ar.a.10044. [DOI] [PubMed] [Google Scholar]
  • 40.Hamrick MW, Pennington C, Byron CD. Bone architecture and disc degeneration in the lumbar spine of mice lacking GDF-8 (myostatin). J Orthop Res. 2003;21(6):1025–32. doi: 10.1016/S0736-0266(03)00105-0. [DOI] [PubMed] [Google Scholar]
  • 41.Lang DH, Conroy DE, Lionikas A, Mack HA, Larsson L, Vogler GP, Vandenbergh DJ, Blizard DA, McClearn GE, Sharkey NA. Bone, muscle, and physical activity: structural equation modeling of relationships and genetic influence with age. J Bone Miner Res. 2009;24(9):1608–17. doi: 10.1359/JBMR.090418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Christoforidis A, Maniadaki I, Stanhope R. Growth hormone/insulin-like growth factor-1 axis during puberty. Pediatr Endocrinol Rev. 2005;3(1):5–10. [PubMed] [Google Scholar]
  • 43.Rauch F, Bailey DA, Baxter-Jones A, Mirwald R, Faulkner R. The “muscle-bone unit” during the pubertal growth spurt. Bone. 2004;34(5):771–5. doi: 10.1016/j.bone.2004.01.022. [DOI] [PubMed] [Google Scholar]
  • 44.Rittweger J, Beller G, Ehrig J, Jung C, Koch U, Ramolla J, Schmidt F, Newitt D, Majumdar S, Schiessl H, Felsenberg D. Bone-muscle strength indices for the human lower leg. Bone. 2000;27(2):319–26. doi: 10.1016/s8756-3282(00)00327-6. [DOI] [PubMed] [Google Scholar]
  • 45.Riggs BL, Khosla S, Melton LJ., 3rd Sex steroids and the construction and conservation of the adult skeleton. Endocr Rev. 2002;23(3):279–302. doi: 10.1210/edrv.23.3.0465. [DOI] [PubMed] [Google Scholar]
  • 46.Bachrach LK. Chapter 14. Skeletal development in childhood and adolescence. In: Rosen CJ, Compston JE, Lian JB, editors. Primer on the metabolic bone diseases and disorders of mineral metabolism. 7th ed. John Wiley & Sons, Inc.; Hoboken, NJ, USA: 2009. pp. 74–9. [Google Scholar]
  • 47.Seeman E. Clinical review 137: sexual dimorphism in skeletal size, density, and strength. J Clin Endocrinol Metab. 2001;86(10):4576–84. doi: 10.1210/jcem.86.10.7960. [DOI] [PubMed] [Google Scholar]
  • 48.Venken K, Callewaert F, Boonen S, Vanderschueren D. Sex hormones, their receptors and bone health. Osteoporos Int. 2008;19(11):1517–25. doi: 10.1007/s00198-008-0609-z. [DOI] [PubMed] [Google Scholar]
  • 49.MacLean HE, Chiu WS, Notini AJ, Axell AM, Davey RA, McManus JF, Ma C, Plant DR, Lynch GS, Zajac JD. Impaired skeletal muscle development and function in male, but not female, genomic androgen receptor knockout mice. FASEB J. 2008;22(8):2676–89. doi: 10.1096/fj.08-105726. [DOI] [PubMed] [Google Scholar]
  • 50.Maughan RJ, Watson JS, Weir J. Strength and cross-sectional area of human skeletal muscle. J Physiol. 1983;338:37–49. doi: 10.1113/jphysiol.1983.sp014658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Ferrando AA, Sheffield-Moore M, Paddon-Jones D, Wolfe RR, Urban RJ. Differential anabolic effects of testosterone and amino acid feeding in older men. J Clin Endocrinol Metab. 2003;88(1):358–62. doi: 10.1210/jc.2002-021041. [DOI] [PubMed] [Google Scholar]
  • 52.Axell AM, MacLean HE, Plant DR, Harcourt LJ, Davis JA, Jimenez M, Handelsman DJ, Lynch GS, Zajac JD. Continuous testosterone administration prevents skeletal muscle atrophy and enhances resistance to fatigue in orchidectomized male mice. Am J Physiol Endocrinol Metab. 2006;291(3):E506–16. doi: 10.1152/ajpendo.00058.2006. [DOI] [PubMed] [Google Scholar]
  • 53.Taaffe DR, Newman AB, Haggerty CL, Colbert LH, de Rekeneire N, Visser M, Goodpaster BH, Nevitt MC, Tylavsky FA, Harris TB. Estrogen replacement, muscle composition, and physical function: the Health ABC Study. Med Sci Sports Exerc. 2005;37(10):1741–7. doi: 10.1249/01.mss.0000181678.28092.31. [DOI] [PubMed] [Google Scholar]
  • 54.Kenny AM, Dawson L, Kleppinger A, Iannuzzi-Sucich M, Judge JO. Prevalence of sarcopenia and predictors of skeletal muscle mass in nonobese women who are long-term users of estrogen-replacement therapy. J Gerontol A Biol Sci Med Sci. 2003;58(5):M436–40. doi: 10.1093/gerona/58.5.m436. [DOI] [PubMed] [Google Scholar]
  • 55.Kontulainen S, Kannus P, Haapasalo H, Sievanen H, Pasanen M, Heinonen A, Oja P, Vuori I. Good maintenance of exercise-induced bone gain with decreased training of female tennis and squash players: a prospective 5-year follow-up study of young and old starters and controls. J Bone Miner Res. 2001;16(2):195–201. doi: 10.1359/jbmr.2001.16.2.195. [DOI] [PubMed] [Google Scholar]
  • 56.Haapasalo H, Kontulainen S, Sievanen H, Kannus P, Jarvinen M, Vuori I. Exercise-induced bone gain is due to enlargement in bone size without a change in volumetric bone density: a peripheral quantitative computed tomography study of the upper arms of male tennis players. Bone. 2000;27(3):351–7. doi: 10.1016/s8756-3282(00)00331-8. [DOI] [PubMed] [Google Scholar]
  • 57.Jones HH, Priest JD, Hayes WC, Tichenor CC, Nagel DA. Humeral hypertrophy in response to exercise. J Bone Joint Surg Am. 1977;59(2):204–8. [PubMed] [Google Scholar]
  • 58.Gilsanz V, Wren TA, Sanchez M, Dorey F, Judex S, Rubin C. Low-level, high-frequency mechanical signals enhance musculoskeletal development of young women with low BMD. J Bone Miner Res. 2006;21(9):1464–74. doi: 10.1359/jbmr.060612. [DOI] [PubMed] [Google Scholar]
  • 59.Kesavan C, Baylink DJ, Kapoor S, Mohan S. Novel loci regulating bone anabolic response to loading: expression QTL analysis in C57BL/6JXC3H/HeJ mice cross. Bone. 2007;41(2):223–30. doi: 10.1016/j.bone.2007.04.185. [DOI] [PubMed] [Google Scholar]
  • 60.Vernikos J, Schneider VS. Space, gravity and the physiology of aging: parallel or convergent disciplines? A mini-review. Gerontology. 2010;56(2):157–66. doi: 10.1159/000252852. [DOI] [PubMed] [Google Scholar]
  • 61.Tasdemir HA, Buyukavci M, Akcay F, Polat P, Yildiran A, Karakelleoglu C. Bone mineral density in children with cerebral palsy. Pediatr Int. 2001;43(2):157–60. doi: 10.1046/j.1442-200x.2001.01352.x. [DOI] [PubMed] [Google Scholar]
  • 62.Shaw NJ, White CP, Fraser WD, Rosenbloom L. Osteopenia in cerebral palsy. Arch Dis Child. 1994;71(3):235–8. doi: 10.1136/adc.71.3.235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Hsu JD. Skeletal changes in children with neuromuscular disorders. Prog Clin Biol Res. 1982;101:553–7. [PubMed] [Google Scholar]
  • 64.Larson CM, Henderson RC. Bone mineral density and fractures in boys with Duchenne muscular dystrophy. J Pediatr Orthop. 2000;20(1):71–4. [PubMed] [Google Scholar]
  • 65.Frey-Rindova P, de Bruin ED, Stussi E, Dambacher MA, Dietz V. Bone mineral density in upper and lower extremities during 12 months after spinal cord injury measured by peripheral quantitative computed tomography. Spinal Cord. 2000;38(1):26–32. doi: 10.1038/sj.sc.3100905. [DOI] [PubMed] [Google Scholar]
  • 66.Castro MJ, Apple DF, Jr, Staron RS, Campos GE, Dudley GA. Influence of complete spinal cord injury on skeletal muscle within 6 mo of injury. J Appl Physiol. 1999;86(1):350–8. doi: 10.1152/jappl.1999.86.1.350. [DOI] [PubMed] [Google Scholar]
  • 67.Richardson CA. The health of the human skeletal system for weight bearing against gravity: The role of deloading the musculo-skeletal system in the development of musculoskeletal injury. J Gravit Physiol. 2002;9(1):P7–10. [PubMed] [Google Scholar]
  • 68.van Ingen Schenau GJ, Boots PJ, de Groot G, Snackers RJ, van Woensel WW. The constrained control of force and position in multi-joint movements. Neuroscience. 1992;46(1):197–207. doi: 10.1016/0306-4522(92)90019-x. [DOI] [PubMed] [Google Scholar]
  • 69.Richardson C, Bullock MI. Changes in muscle activity during fast, alternating flexion-extension movements of the knee. Scand J Rehabil Med. 1986;18(2):51–8. [PubMed] [Google Scholar]
  • 70.Hodges PW, Richardson CA. The influence of isometric hip adduction on quadriceps femoris activity. Scand J Rehabil Med. 1993;25(2):57–62. [PubMed] [Google Scholar]
  • 71.Fitts RH, Riley DR, Widrick JJ. Functional and structural adaptations of skeletal muscle to microgravity. J Exp Biol. 2001;204(Pt 18):3201–8. doi: 10.1242/jeb.204.18.3201. [DOI] [PubMed] [Google Scholar]
  • 72.Fitts RH, Riley DR, Widrick JJ. Physiology of a microgravity environment invited review: microgravity and skeletal muscle. J Appl Physiol. 2000;89(2):823–39. doi: 10.1152/jappl.2000.89.2.823. [DOI] [PubMed] [Google Scholar]
  • 73.Edgerton VR, Zhou MY, Ohira Y, Klitgaard H, Jiang B, Bell G, Harris B, Saltin B, Gollnick PD, Roy RR, et al. Human fiber size and enzymatic properties after 5 and 11 days of spaceflight. J Appl Physiol. 1995;78(5):1733–9. doi: 10.1152/jappl.1995.78.5.1733. [DOI] [PubMed] [Google Scholar]
  • 74.Belavy DL, Richardson CA, Wilson SJ, Felsenberg D, Rittweger J. Tonic-to-phasic shift of lumbo-pelvic muscle activity during 8 weeks of bed rest and 6-months follow up. J Appl Physiol. 2007;103(1):48–54. doi: 10.1152/japplphysiol.00850.2006. [DOI] [PubMed] [Google Scholar]
  • 75.Andersen JL. Muscle fibre type adaptation in the elderly human muscle. Scand J Med Sci Sports. 2003;13(1):40–7. doi: 10.1034/j.1600-0838.2003.00299.x. [DOI] [PubMed] [Google Scholar]
  • 76.Roland M, Hanson AM, Cannon CM, Stodieck LS, Ferguson VL. Exercise prevention of unloading-induced bone and muscle loss in adult mice. Biomed Sci Instrum. 2005;41:128–34. [PubMed] [Google Scholar]
  • 77.Amblard D, Lafage-Proust MH, Laib A, Thomas T, Ruegsegger P, Alexandre C, Vico L. Tail suspension induces bone loss in skeletally mature mice in the C57BL/6J strain but not in the C3H/HeJ strain. J Bone Miner Res. 2003;18(3):561–9. doi: 10.1359/jbmr.2003.18.3.561. [DOI] [PubMed] [Google Scholar]
  • 78.Goodpaster BH, Park SW, Harris TB, Kritchevsky SB, Nevitt M, Schwartz AV, Simonsick EM, Tylavsky FA, Visser M, Newman AB. The loss of skeletal muscle strength, mass, and quality in older adults: the health, aging and body composition study. J Gerontol A Biol Sci Med Sci. 2006;61(10):1059–64. doi: 10.1093/gerona/61.10.1059. [DOI] [PubMed] [Google Scholar]
  • 79.Tichet J, Vol S, Goxe D, Salle A, Berrut G, Ritz P. Prevalence of sarcopenia in the French senior population. J Nutr Health Aging. 2008;12(3):202–6. doi: 10.1007/BF02982621. [DOI] [PubMed] [Google Scholar]
  • 80.Lauretani F, Bandinelli S, Bartali B, Di Iorio A, Giacomini V, Corsi AM, Guralnik JM, Ferrucci L. Axonal degeneration affects muscle density in older men and women. Neurobiol Aging. 2006;27(8):1145–54. doi: 10.1016/j.neurobiolaging.2005.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Fox KM, Magaziner J, Hawkes WG, Yu-Yahiro J, Hebel JR, Zimmerman SI, Holder L, Michael R. Loss of bone density and lean body mass after hip fracture. Osteoporos Int. 2000;11(1):31–5. doi: 10.1007/s001980050003. [DOI] [PubMed] [Google Scholar]
  • 82.Alley DE, Koster A, Mackey D, Cawthon P, Ferrucci L, Simonsick EM, Yu B, Hardy S, Goodpaster B, Sarkisian C, Houston DK, Kritchevsky SB, Cummings S, Lee JS, Tylavsky FA, Newman A, Harris T. Hospitalization and change in body composition and strength in a population-based cohort of older persons. J Am Geriatr Soc. 2010;58(11):2085–91. doi: 10.1111/j.1532-5415.2010.03144.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Kortebein P, Symons TB, Ferrando A, Paddon-Jones D, Ronsen O, Protas E, Conger S, Lombeida J, Wolfe R, Evans WJ. Functional impact of 10 days of bed rest in healthy older adults. J Gerontol A Biol Sci Med Sci. 2008;63(10):1076–81. doi: 10.1093/gerona/63.10.1076. [DOI] [PubMed] [Google Scholar]
  • 84.Hagen JL, Krause DJ, Baker DJ, Fu MH, Tarnopolsky MA, Hepple RT. Skeletal muscle aging in F344BN F1-hybrid rats: I. Mitochondrial dysfunction contributes to the age-associated reduction in VO2max. J Gerontol A Biol Sci Med Sci. 2004;59(11):1099–110. doi: 10.1093/gerona/59.11.1099. [DOI] [PubMed] [Google Scholar]
  • 85.Cree MG, Newcomer BR, Katsanos CS, Sheffield-Moore M, Chinkes D, Aarsland A, Urban R, Wolfe RR. Intramuscular and liver triglycerides are increased in the elderly. J Clin Endocrinol Metab. 2004;89(8):3864–71. doi: 10.1210/jc.2003-031986. [DOI] [PubMed] [Google Scholar]
  • 86.Dela F, Kjaer M. Resistance training, insulin sensitivity and muscle function in the elderly. Essays Biochem. 2006;42:75–88. doi: 10.1042/bse0420075. [DOI] [PubMed] [Google Scholar]
  • 87.Bonaldo P, Sandri M. Cellular and molecular mechanisms of muscle atrophy. Dis Model Mech. 2013;6(1):25–39. doi: 10.1242/dmm.010389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Mizushima N, Levine B, Cuervo AM, Klionsky DJ. Autophagy fights disease through cellular self-digestion. Nature. 2008;451(7182):1069–75. doi: 10.1038/nature06639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Carmignac V, Svensson M, Korner Z, Elowsson L, Matsumura C, Gawlik KI, Allamand V, Durbeej M. Autophagy is increased in laminin alpha2 chain-deficient muscle and its inhibition improves muscle morphology in a mouse model of MDC1A. Hum Mol Genet. 2011;20(24):4891–902. doi: 10.1093/hmg/ddr427. [DOI] [PubMed] [Google Scholar]
  • 90.Malicdan MC, Noguchi S, Nonaka I, Saftig P, Nishino I. Lysosomal myopathies: an excessive build-up in autophagosomes is too much to handle. Neuromuscul Disord. 2008;18(7):521–9. doi: 10.1016/j.nmd.2008.04.010. [DOI] [PubMed] [Google Scholar]
  • 91.Cuervo AM. Autophagy and aging: keeping that old broom working. Trends Genet. 2008;24(12):604–12. doi: 10.1016/j.tig.2008.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Wenz T, Rossi SG, Rotundo RL, Spiegelman BM, Moraes CT. Increased muscle PGC-1alpha expression protects from sarcopenia and metabolic disease during aging. Proc Natl Acad Sci U S A. 2009;106(48):20405–10. doi: 10.1073/pnas.0911570106. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 93.Jang YC, Sinha M, Cerletti M, Dall'Osso C, Wagers AJ. Skeletal muscle stem cells: effects of aging and metabolism on muscle regenerative function. Cold Spring Harb Symp Quant Biol. 2011;76:101–11. doi: 10.1101/sqb.2011.76.010652. [DOI] [PubMed] [Google Scholar]
  • 94.Tothova Z, Kollipara R, Huntly BJ, Lee BH, Castrillon DH, Cullen DE, McDowell EP, Lazo-Kallanian S, Williams IR, Sears C, Armstrong SA, Passegue E, DePinho RA, Gilliland DG. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell. 2007;128(2):325–39. doi: 10.1016/j.cell.2007.01.003. [DOI] [PubMed] [Google Scholar]
  • 95.Renault VM, Rafalski VA, Morgan AA, Salih DA, Brett JO, Webb AE, Villeda SA, Thekkat PU, Guillerey C, Denko NC, Palmer TD, Butte AJ, Brunet A. FoxO3 regulates neural stem cell homeostasis. Cell Stem Cell. 2009;5(5):527–39. doi: 10.1016/j.stem.2009.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Carlson BM, Faulkner JA. Muscle transplantation between young and old rats: age of host determines recovery. Am J Physiol. 1989;256(6 Pt 1):C1262–6. doi: 10.1152/ajpcell.1989.256.6.C1262. [DOI] [PubMed] [Google Scholar]
  • 97.Conboy IM, Conboy MJ, Wagers AJ, Girma ER, Weissman IL, Rando TA. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature. 2005;433(7027):760–4. doi: 10.1038/nature03260. [DOI] [PubMed] [Google Scholar]
  • 98.Conboy IM, Conboy MJ, Smythe GM, Rando TA. Notch-mediated restoration of regenerative potential to aged muscle. Science. 2003;302(5650):1575–7. doi: 10.1126/science.1087573. [DOI] [PubMed] [Google Scholar]
  • 99.Brack AS, Conboy MJ, Roy S, Lee M, Kuo CJ, Keller C, Rando TA. Increased Wnt signaling during aging alters muscle stem cell fate and increases fibrosis. Science. 2007;317(5839):807–10. doi: 10.1126/science.1144090. [DOI] [PubMed] [Google Scholar]
  • 100.Kassem M, Marie PJ. Senescence-associated intrinsic mechanisms of osteoblast dysfunctions. Aging Cell. 2011;10(2):191–7. doi: 10.1111/j.1474-9726.2011.00669.x. [DOI] [PubMed] [Google Scholar]
  • 101.Boonen S, Mohan S, Dequeker J, Aerssens J, Vanderschueren D, Verbeke G, Broos P, Bouillon R, Baylink DJ. Down-regulation of the serum stimulatory components of the insulin-like growth factor (IGF) system (IGF-I, IGF-II, IGF binding protein [BP]-3, and IGFBP-5) in age-related (type II) femoral neck osteoporosis. J Bone Miner Res. 1999;14(12):2150–8. doi: 10.1359/jbmr.1999.14.12.2150. [DOI] [PubMed] [Google Scholar]
  • 102.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(4):701–4. doi: 10.1210/jcem-59-4-701. [DOI] [PubMed] [Google Scholar]
  • 103.Ho KY, Evans WS, Blizzard RM, Veldhuis JD, Merriam GR, Samojlik E, Furlanetto R, Rogol AD, Kaiser DL, Thorner MO. Effects of sex and age on the 24-hour profile of growth hormone secretion in man: importance of endogenous estradiol concentrations. J Clin Endocrinol Metab. 1987;64(1):51–8. doi: 10.1210/jcem-64-1-51. [DOI] [PubMed] [Google Scholar]
  • 104.Kousteni S. FoxOs: unifying links between oxidative stress and skeletal homeostasis. Curr Osteoporos Rep. 2011;9(2):60–6. doi: 10.1007/s11914-011-0054-3. [DOI] [PubMed] [Google Scholar]
  • 105.Sun Y, Li W, Lu Z, Chen R, Ling J, Ran Q, Jilka RL, Chen XD. Rescuing replication and osteogenesis of aged mesenchymal stem cells by exposure to a young extracellular matrix. FASEB J. 2011;25(5):1474–85. doi: 10.1096/fj.10-161497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Garnero P, Sornay-Rendu E, Chapuy MC, Delmas PD. Increased bone turnover in late postmenopausal women is a major determinant of osteoporosis. J Bone Miner Res. 1996;11(3):337–49. doi: 10.1002/jbmr.5650110307. [DOI] [PubMed] [Google Scholar]
  • 107.Pearson OM, Lieberman DE. The aging of Wolff's “law”: ontogeny and responses to mechanical loading in cortical bone. Am J Phys Anthropol Suppl. 2004;39:63–99. doi: 10.1002/ajpa.20155. [DOI] [PubMed] [Google Scholar]
  • 108.Song MY, Ruts E, Kim J, Janumala I, Heymsfield S, Gallagher D. Sarcopenia and increased adipose tissue infiltration of muscle in elderly African American women. Am J Clin Nutr. 2004;79(5):874–80. doi: 10.1093/ajcn/79.5.874. [DOI] [PubMed] [Google Scholar]
  • 109.Rolland Y, Lauwers-Cances V, Cristini C, Abellan van Kan G, Janssen I, Morley JE, Vellas B. Difficulties with physical function associated with obesity, sarcopenia, and sarcopenic-obesity in community-dwelling elderly women: the EPIDOS (EPIDemiologie de l'OSteoporose) Study. Am J Clin Nutr. 2009;89(6):1895–900. doi: 10.3945/ajcn.2008.26950. [DOI] [PubMed] [Google Scholar]
  • 110.Beasley LE, Koster A, Newman AB, Javaid MK, Ferrucci L, Kritchevsky SB, Kuller LH, Pahor M, Schaap LA, Visser M, Rubin SM, Goodpaster BH, Harris TB. Inflammation and race and gender differences in computerized tomography-measured adipose depots. Obesity (Silver Spring. 2009;17(5):1062–9. doi: 10.1038/oby.2008.627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Hsu YH, Fox CS, Bouxsein ML, Hoffmann U, Cupples LA, Kiel DP. Peri-aortic fat is negatively associated with volumetric bone density, cross-sectional area and compressive strength of lumbar vertebrae: the Framingham Osteoporosis Study [Abstract] [Internet]. [2013 Apr 22];J Bone Miner Res. 2011 26(Suppl 1):S1. Available from: http://www.asbmr.org/Meetings/AnnualMeeting/AbstractDetail.aspx?aid=34563ced-7f95-4691-9632-867f03f970ba. [Google Scholar]
  • 112.Pedersen BK, Febbraio MA. Muscles, exercise and obesity: skeletal muscle as a secretory organ. Nat Rev Endocrinol. 2012;8(8):457–65. doi: 10.1038/nrendo.2012.49. [DOI] [PubMed] [Google Scholar]
  • 113.Ellingsgaard H, Hauselmann I, Schuler B, Habib AM, Baggio LL, Meier DT, Eppler E, Bouzakri K, Wueest S, Muller YD, Hansen AM, Reinecke M, Konrad D, Gassmann M, Reimann F, Halban PA, Gromada J, Drucker DJ, Gribble FM, Ehses JA, Donath MY. Interleukin-6 enhances insulin secretion by increasing glucagon-like peptide-1 secretion from L cells and alpha cells. Nat Med. 2011;17(11):1481–9. doi: 10.1038/nm.2513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Bostrom P, Wu J, Jedrychowski MP, Korde A, Ye L, Lo JC, Rasbach KA, Bostrom EA, Choi JH, Long JZ, Kajimura S, Zingaretti MC, Vind BF, Tu H, Cinti S, Hojlund K, Gygi SP, Spiegelman BM. A PGC1-alpha-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature. 2012;481(7382):463–8. doi: 10.1038/nature10777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Kawai M, de Paula FJ, Rosen CJ. New insights into osteoporosis: the bone-fat connection. J Intern Med. 2012;272(4):317–29. doi: 10.1111/j.1365-2796.2012.02564.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Ponrartana S, Aggabao PC, Hu HH, Aldrovandi GM, Wren TA, Gilsanz V. Brown adipose tissue and its relationship to bone structure in pediatric patients. J Clin Endocrinol Metab. 2012;97(8):2693–8. doi: 10.1210/jc.2012-1589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Yasui A, Nishizawa H, Okuno Y, Morita K, Kobayashi H, Kawai K, Matsuda M, Kishida K, Kihara S, Kamei Y, Ogawa Y, Funahashi T, Shimomura I. Foxo1 represses expression of musclin, a skeletal muscle-derived secretory factor. Biochem Biophys Res Commun. 2007;364(2):358–65. doi: 10.1016/j.bbrc.2007.10.013. [DOI] [PubMed] [Google Scholar]
  • 118.Moffatt P, Thomas GP. Osteocrin—beyond just another bone protein?. Cell Mol Life Sci. 2009;66(7):1135–9. doi: 10.1007/s00018-009-8716-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Bogdanovich S, Krag TO, Barton ER, Morris LD, Whittemore LA, Ahima RS, Khurana TS. Functional improvement of dystrophic muscle by myostatin blockade. Nature. 2002;420(6914):418–21. doi: 10.1038/nature01154. [DOI] [PubMed] [Google Scholar]
  • 120.Whittemore LA, Song K, Li X, Aghajanian J, Davies M, Girgenrath S, Hill JJ, Jalenak M, Kelley P, Knight A, Maylor R, O'Hara D, Pearson A, Quazi A, Ryerson S, Tan XY, Tomkinson KN, Veldman GM, Widom A, Wright JF, Wudyka S, Zhao L, Wolfman NM. Inhibition of myostatin in adult mice increases skeletal muscle mass and strength. Biochem Biophys Res Commun. 2003;300(4):965–71. doi: 10.1016/s0006-291x(02)02953-4. [DOI] [PubMed] [Google Scholar]
  • 121.Wolfman NM, McPherron AC, Pappano WN, Davies MV, Song K, Tomkinson KN, Wright JF, Zhao L, Sebald SM, Greenspan DS, Lee SJ. Activation of latent myostatin by the BMP-1/tolloid family of metalloproteinases. Proc Natl Acad Sci U S A. 2003;100(26):15842–6. doi: 10.1073/pnas.2534946100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Lee SJ, Reed LA, Davies MV, Girgenrath S, Goad ME, Tomkinson KN, Wright JF, Barker C, Ehrmantraut G, Holmstrom J, Trowell B, Gertz B, Jiang MS, Sebald SM, Matzuk M, Li E, Liang LF, Quattlebaum E, Stotish RL, Wolfman NM. Regulation of muscle growth by multiple ligands signaling through activin type II receptors. Proc Natl Acad Sci U S A. 2005;102(50):18117–22. doi: 10.1073/pnas.0505996102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Pearsall RS, Canalis E, Cornwall-Brady M, Underwood KW, Haigis B, Ucran J, Kumar R, Pobre E, Grinberg A, Werner ED, Glatt V, Stadmeyer L, Smith D, Seehra J, Bouxsein ML. A soluble activin type IIA receptor induces bone formation and improves skeletal integrity. Proc Natl Acad Sci U S A. 2008;105(19):7082–7. doi: 10.1073/pnas.0711263105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Fajardo RJ, Manoharan RK, Pearsall RS, Davies MV, Marvell T, Monnell TE, Ucran JA, Pearsall AE, Khanzode D, Kumar R, Underwood KW, Roberts B, Seehra J, Bouxsein ML. Treatment with a soluble receptor for activin improves bone mass and structure in the axial and appendicular skeleton of female cynomolgus macaques (Macaca fascicularis). Bone. 2010;46(1):64–71. doi: 10.1016/j.bone.2009.09.018. [DOI] [PubMed] [Google Scholar]
  • 125.Lotinun S, Pearsall RS, Davies MV, Marvell TH, Monnell TE, Ucran J, Fajardo RJ, Kumar R, Underwood KW, Seehra J, Bouxsein ML, Baron R. A soluble activin receptor Type IIA fusion protein (ACE-011) increases bone mass via a dual anabolic-antiresorptive effect in Cynomolgus monkeys. Bone. 2010;46(4):1082–8. doi: 10.1016/j.bone.2010.01.370. [DOI] [PubMed] [Google Scholar]
  • 126.Chantry AD, Heath D, Mulivor AW, Pearsall S, Baud'huin M, Coulton L, Evans H, Abdul N, Werner ED, Bouxsein ML, Key ML, Seehra J, Arnett TR, Vanderkerken K, Croucher P. Inhibiting activin-A signaling stimulates bone formation and prevents cancer-induced bone destruction in vivo. J Bone Miner Res. 2010;25(12):2633–46. doi: 10.1002/jbmr.142. [DOI] [PubMed] [Google Scholar]
  • 127.Koncarevic A, Cornwall-Brady M, Pullen A, Davies M, Sako D, Liu J, Kumar R, Tomkinson K, Baker T, Umiker B, Monnell T, Grinberg AV, Liharska K, Underwood KW, Ucran JA, Howard E, Barberio J, Spaits M, Pearsall S, Seehra J, Lachey J. A soluble activin receptor type IIb prevents the effects of androgen deprivation on body composition and bone health. Endocrinology. 2010;151(9):4289–300. doi: 10.1210/en.2010-0134. [DOI] [PubMed] [Google Scholar]
  • 128.Vallet S, Mukherjee S, Vaghela N, Hideshima T, Fulciniti M, Pozzi S, Santo L, Cirstea D, Patel K, Sohani AR, Guimaraes A, Xie W, Chauhan D, Schoonmaker JA, Attar E, Churchill M, Weller E, Munshi N, Seehra JS, Weissleder R, Anderson KC, Scadden DT, Raje N. Activin A promotes multiple myeloma-induced osteolysis and is a promising target for myeloma bone disease. Proc Natl Acad Sci U S A. 2010;107(11):5124–9. doi: 10.1073/pnas.0911929107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.DiGirolamo DJ, Singhal V, Clemens TL, Lee SJ. Systemic administration of soluble activin receptors produces differential anabolic effects in muscle and bone in mice [Abstract] [Internet]. [2013 Apr 22];J Bone Miner Res. 2011 2011 26(Suppl 1):S1. Available from: http://www.asbmr.org/Meetings/AnnualMeeting/AbstractDetail.aspx?aid=27458272-83b7-4749-8f0c-e08f31d17152. [Google Scholar]
  • 130.Frost HM. Bone's mechanostat: a 2003 update. Anat Rec A Discov Mol Cell Evol Biol. 2003;275(2):1081–101. doi: 10.1002/ar.a.10119. [DOI] [PubMed] [Google Scholar]
  • 131.Lewis SJ, Smith PE. Osteoporosis prevention in myasthenia gravis: a reminder. Acta Neurol Scand. 2001;103(5):320–2. doi: 10.1034/j.1600-0404.2001.103005320.x. [DOI] [PubMed] [Google Scholar]
  • 132.Schiaffino S, Sandri M, Murgia M. Activity-dependent signaling pathways controlling muscle diversity and plasticity. Physiology (Bethesda) 2007;22:269–78. doi: 10.1152/physiol.00009.2007. [DOI] [PubMed] [Google Scholar]
  • 133.Takeda S, Karsenty G. Molecular bases of the sympathetic regulation of bone mass. Bone. 2008;42(5):837–40. doi: 10.1016/j.bone.2008.01.005. [DOI] [PubMed] [Google Scholar]
  • 134.Yirmiya R, Goshen I, Bajayo A, Kreisel T, Feldman S, Tam J, Trembovler V, Csernus V, Shohami E, Bab I. Depression induces bone loss through stimulation of the sympathetic nervous system. Proc Natl Acad Sci U S A. 2006;103(45):16876–81. doi: 10.1073/pnas.0604234103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Elefteriou F, Ahn JD, Takeda S, Starbuck M, Yang X, Liu X, Kondo H, Richards WG, Bannon TW, Noda M, Clement K, Vaisse C, Karsenty G. Leptin regulation of bone resorption by the sympathetic nervous system and CART. Nature. 2005;434(7032):514–20. doi: 10.1038/nature03398. [DOI] [PubMed] [Google Scholar]

RESOURCES