Вы находитесь на странице: 1из 22

REVIEW

Effects of Leptin on the Skeleton

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


Ian R. Reid,1,2 Paul A Baldock,3 and Jillian Cornish1

1
University of Auckland, Auckland 1132, New Zealand; 2Department of Endocrinology, Auckland District
Health Board, Auckland 1132, New Zealand; and 3Garvan Institute of Medical Research, Sydney, New South
Wales 2010, Australia
ORCiD numbers: 0000-0001-6021-5458 (I. R. Reid).

ABSTRACT Leptin originates in adipocytes, including those in bone marrow, and circulates in concentrations 20 to 90 times higher
than those in the cerebrospinal fluid. It has direct anabolic effects on osteoblasts and chondrocytes, but it also influences bone
indirectly, via the hypothalamus and sympathetic nervous system, via changes in body weight, and via effects on the production of
other hormones (e.g., pituitary). Leptin’s role in bone physiology is determined by the balance of these conflicting effects. Reflecting
this inconsistency, the leptin-deficient mouse has reduced length and bone mineral content of long bones but increased vertebral
trabecular bone. A consistent bone phenotype in human leptin deficiency has not been established. Systemic leptin administration in
animals and humans usually exerts a positive effect on bone mass, and leptin administration into the cerebral ventricles usually
normalizes the bone phenotype in leptin-deficient mice. Reflecting the role of the sympathetic nervous system in mediating the central
catabolic effects of leptin on the skeleton, b-adrenergic agonists and antagonists have major effects on bone in mice, but this is not
consistently seen in humans. The balance of the central and peripheral effects of leptin on bone remains an area of substantial
controversy and might vary between species and according to other factors such as body weight, baseline circulating leptin levels, and
the presence of specific pathologies. In humans, leptin is likely to contribute to the positive relationship observed between adiposity
and bone density, which allows the skeleton to respond appropriately to changes in soft tissue mass. (Endocrine Reviews 39: 938 – 959,
2018)

W hen the adipokine leptin was discovered in


the s, there was a growing awareness
of the importance of body weight in determining
effects on bone of leptin administration into the
central nervous system (CNS). There is apparent
disagreement between these bodies of research,
bone density and risk of fracture. That area of with leptin being found to be anabolic and
clinical research has expanded considerably in the antiresorptive when applied directly to bone cells
last two decades, with the critical role of fat mass on and tissue, whereas its central administration
bone density becoming established. Body weight is in mice results in substantial bone loss at some
now an integral part of the routine clinical sites. Administration of leptin affects many fac-
evaluation of fracture risk, being used in the major tors that can influence bone metabolism, such as
ISSN Print: 0163-769X risk calculators such as FRAX, Garvan, and body weight and sex hormone production, which
ISSN Online: 1945-7189 QFracture. complicates interpretation of experimental data.
Printed: in USA The clinical recognition of an important role of Understanding the fat-bone connection is further
Copyright © 2018
adipose tissue in bone health led to an explora- complicated by studies of leptin-deficient animal
Endocrine Society
Received: 11 October 2017
tion of the effects of a key adipokine, leptin, on models, which have the leptin levels of starvation but
Accepted: 26 June 2018 bone physiology. This literature can be parti- display many other features of obesity. These bodies
First Published Online: tioned into studies of the direct effects of leptin on of work will be presented to elucidate how these
31 August 2018 bone cells and tissue and studies of the indirect conflicting effects play out in intact animals and in

938 https://academic.oup.com/edrv doi: 10.1210/er.2017-00226


REVIEW

ESSENTIAL POINTS
· Leptin originates in adipose tissue, including in bone marrow adipocytes, and acts directly on osteoblasts and
chondrocytes to increase their growth
· Whereas the leptin-deficient mouse (ob/ob) has reduced length and bone mineral content of long bones but increased
trabecular bone volume in the vertebrae, a consistent bone phenotype in human leptin deficiency has not been
established
· Systemic leptin administration in animals and humans almost always exerts a positive effect on bone mass, except when
given in very high dosages, which substantially reduce body weight as a result of leptin’s anorectic effects
··

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


Leptin administration into the third cerebral ventricle usually normalizes the bone phenotype in leptin-deficient animals
The balance of the central and peripheral effects of leptin on bone remains an area of controversy and might vary between
species
· In humans, leptin is likely to contribute to the positive relationship observed between adiposity and bone density

human health and disease. To place these descriptions in current understanding of how soft tissue and bone health
the context of human physiology it is useful to review the interact in humans.

Clinical Impact of Soft Tissue on Bone Health that the effect of BMI is mediated through its effect on
BMD. In contrast, for hip fracture there is a residual
Bone density protective effect from high BMI even after adjustment
Many studies have demonstrated that bone mineral for BMD, possibly mediated by the shock-absorbing
density (BMD) is positively related to indices of soft effect of the adipose tissue over the greater trochanter.
tissue mass, whether these be body weight, body mass Some studies have related fracture risk to fat and
index (BMI), fat mass, or lean mass () (Fig. ). Con- lean masses separately. The Study of Osteoporotic
troversy has existed as to whether fat or lean masses exert Fractures found that both lean mass and fat mass were
the greater effect on BMD, and that may depend on age, inversely related to hip fracture risk (). A French
sex, gonadal status, and habitual exercise levels (). Some study observed a % increase in hip fracture risk for
authors have used statistical techniques to tease apart the each standard deviation decrease in fat mass but no
interacting effects of fat and lean tissue on BMD. This is effect of lean mass (). A study of Chinese men in
challenging because of their significant intercorrelation Hong Kong found that the OR for vertebral fracture
and because of the even stronger correlations between fat was . when the first quartile of fat mass was com-
mass and body weight. Entering such strongly inter- pared with the fourth quartile, whereas comparisons of
correlated variables as “independent” variables into a the same quartiles for lean mass produced a non-
multiple regression analysis violates the assumptions on significant OR of . (). Thus, the clinical literature
which the analysis is based, and nonsense findings can strongly indicates that adiposity is protective against
result (). This is the likely explanation for the finding in fracture. However, more recent studies suggest that
some studies that fat mass is inversely related to BMD. this effect may vary across fracture sites and that
When entered individually, body weight, lean mass, and fractures of the upper arm and lower leg are more
fat mass are consistently positively related to BMD, common in heavier people (, , ). Fall risk or the
providing a mechanism by which the skeleton is able to skeletal loading at these sites after falls might account
adjust to the load it bears. This explanation is consistent for these apparent inconsistencies.
with the fracture data, which also indicate that adiposity is
protective against fracture.
Leptin Biology
Fracture risk
Epidemiological studies consistently demonstrate that Maintenance of adequate energy stores is a necessity
low body weight is a risk factor for fracture. Meta- for survival. Management of these stores entails the
analyses of data from , to , adults in control of short-term energy balance, by matching
prospective, population-based cohorts show that total food intake to energy use. However, there also exist
fractures, osteoporotic fractures, and hip fractures are long-term considerations in the maintenance of en-
all inversely related to BMI in both men and women ergy reserves to ensure survival over seasonal changes,
(, ) (Fig. ). If fracture risk is adjusted for BMD, the such as altered food availability, requirements for
dependence of risk on BMI is no longer significant for reproduction, and thermoregulatory changes. Failure
total fractures and osteoporotic fractures, suggesting to properly control both short- and long-term energy

doi: 10.1210/er.2017-00226 https://academic.oup.com/edrv 939


REVIEW

Figure 1. Dependence of BMD and fracture risk on BMI in postmenopausal women. The left panel shows the regression relationship
between total hip BMD (in grams per square centimeter) and BMI in 1462 normal postmenopausal women from the Auckland Calcium
Study (1). r is the Pearson correlation coefficient. The right panel shows the relationship between BMI and risk of fracture [hazard ratio
(HR) vs BMI 25 kg/m2] for osteoporotic fracture (solid line) and hip fracture (dashed line), adjusted for age and time since baseline (2).
Data were gathered from prospective cohorts from more than 25 countries; baseline data on BMI were available from 398,610 women
with an average age of 63 (range 20 to 105) years and follow-up of 2.2 million person-years during which 30,280 osteoporotic fractures
(6457 hip fractures) occurred. Left panel, copyright IR Reid, used with permission. Right panel reproduced with permission from
Johansson H, Kanis JA, Oden A, et al. A meta-analysis of the association of fracture risk and BMI in women. J Bone Miner Res 2014; 29 (1):
223-233 (2). [© 2018 Illustration Presentation ENDOCRINE SOCIETY].

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


4.0 Osteoporotic
fracture
Hip fracture
Total hip BMD

3.0
1.0

HR
2.0

1.0

0.5 r=0.39 P < 0.001


0
10 20 30 40 50 10 20 30 40 50
BMI (kg/m2) © 2018 Illustration Presentation BMI (kg/m2)
ENDOCRINE SOCIETY

balance can have serious survival and reproductive with glucocorticoids, catecholamines, insulin, and
consequences. As a result, powerful regulatory path- cytokines ().
ways have evolved to support these fundamental Leptin action is mediated by the long form of the
processes. Given the bodywide control necessary to leptin receptor (LepRb). This is a type I cytokine
coordinate energy balance, it is not surprising that the receptor of the IL- receptor family [see Flak and
brain takes a pivotal role in setting and maintaining Myers () for a recent review]. It is a single
energetic tone and energy reserves, most notably in the membrane–spanning receptor with an –amino acid
form of fat storage. However, for central control to extracellular domain, a –amino acid transmembrane
operate efficiently, feedback on the energy state of the domain, and a –amino acid intracellular domain
body is necessary. In terms of energy reserves, leptin is (). Several short forms of this receptor also exist,
the dominant marker of fat reserves, signaling in an resulting from alternative RNA splicing at the most
endocrine manner from white adipose tissue to C-terminal coding exon, resulting in intracellular
metabotropic neurons within the hypothalamus and domains of differing length and sequence composition
brainstem, principally to ensure maintenance of suf- (). The very short intracellular domains of the short
ficient energy stores. However, leptin also signals di- forms of the receptor render them unable to signal.
rectly to numerous other tissues, creating a regulatory As a result, loss of LepRb alone, as in the db/db mouse,
network capable of responding in a dynamic manner recapitulates the phenotype of the leptin deficient ob/
to both starvation and obesity. ob mouse, and restoration of LepRb alone in mice
The leptin gene was identified . years ago in a lacking all leptin receptor isoforms normalizes phys-
well-characterized genetically obese mouse (). It iology (). The murine and human receptors are
codes a peptide of  amino acids that is principally highly similar in the amino acid sequences of both the
expressed in white adipose tissue, including that of the extracellular (% identity) and intracellular domains
bone marrow (). It has also been demonstrated in (% identity) (). The binding of leptin to this re-
other tissues, including the breast, ovary, placenta, ceptor results in receptor dimerization and activates a
pituitary, skeletal muscle, stomach, lymphoid tissue, wide variety of signaling pathways including JAK,
mesenchymal stem cells, and bone (–). Cir- STAT, and MAPK (Fig. ). The short isoforms of the
culating leptin levels are pulsatile () and show leptin receptor appear to be involved in the transport
a circadian rhythm with the nadir in the mid- of leptin across the blood-brain barrier and do not
afternoon and the peak at midnight. Circulating have a signaling function ().
leptin levels are directly correlated with body fat
mass but are also rapidly responsive to changes in Leptin signaling in the hypothalamus
calorie intake, falling during starvation (). Women The CNS is an important target for leptin action. The
generally have higher levels than men, even after for fat brain is normally protected from circulatory factors by
mass is controlled for (). This difference suggests the blood-brain barrier; however, in several regions in
that sex hormones influence leptin secretion, along the hypothalamus and brainstem, the blood-brain

940 Reid et al Effects of Leptin on the Skeleton Endocrine Reviews, December 2018, 39(6):938–959
REVIEW

barrier is semipermeable. Neurons in these regions are reflects the actions of reduced leptin signaling through
bathed by circulating factors, such as leptin, and ex- acquired leptin resistance. As a result, both these obese
press leptin receptors, enabling a direct response to models (ob/ob and diet-induced obesity) contain aspects
changes in leptin by altering efferent neural outflow of reduced leptin signaling as occurs in starvation,
and the secretion of endocrine factors that regulate which acts to maintain hyperphagia and promote ad-
peripheral tissues such as the gut, pancreas, gonads, ipose accrual but might also modify the skeletal re-
and bone. sponse to varying degrees.
Leptin receptors are highly expressed in the hy-
pothalamus, where signaling has an anorexic effect. Leptin in the circulation and cerebrospinal fluid
This effect results from activation of neurons in the Although the CNS is a major leptin target, leptin

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


arcuate nucleus that produce pro-opiomelanocortin originates in adipose tissue. It is transported intact
and cocaine- and amphetamine-regulated transcript from blood into the CNS by a saturable system ().
(CART) and from inhibition of orexigenic neurons Leptin levels are much higher in blood than in the
that synthesize agouti-related peptide and neuropep- CNS. Caro et al. () reported that leptin concen-
tide Y (NPY). Signals from the arcuate nucleus, a trations in blood are  times higher than those in the
region of blood-brain barrier permeability, project to cerebrospinal fluid in people of normal-weight and 
numerous regions including the paraventricular nu- times higher in people with obesity. Thus, consistent
cleus, dorsomedial hypothalamus, ventromedial hy- with actions of leptin resistance to reduce leptin
pothalamus, and lateral hypothalamus. The lateral transport across the blood-brain barrier in obesity, the
hypothalamus also contains neurons expressing % increase in circulating leptin concentration in
melanin-concentrating hormone, which is decreased obesity results in only a % increase in cerebrospinal
by leptin, resulting in reduced appetite (). Leptin also fluid leptin levels. Nam et al. () found peripheral
acts via the hypothalamus to regulate the sympathetic leptin levels  times higher than cerebrospinal fluid
nervous system (SNS) to increase energy expenditure values in normal subjects and  times higher in
in brown fat by increasing expression of uncoupling
protein  (UCP) (, ). In addition to regulating
energy homeostasis, leptin regulates hypothalamic- Leptin
pituitary function, influencing levels of thyroid hor-
mones, growth hormone, cortisol, and sex steroids. All
of these have important effects on the skeleton. These
pathways are summarized in the upper part of Fig. .
PTP1B 2 JAK ROCK1
Leptin resistance JAK 2 ?
In obesity, circulating leptin levels are elevated. TCPTP AMPK
However, the counter regulatory effects on appetite IRS2
and thermogenesis are not adequate to result in a Socs3 Tyr985 PTPN11
SH2B1
healthy body weight. Clinical trials of leptin admin-
istration in obesity have shown little reduction in Tyr1077 STAT5 PI3K
weight (), despite the significant effects of leptin in ERK
Tyr1138 STAT3
normal weight or, particularly, leptin-deficient ani-
mals. As a result, obesity is regarded in part as a state of
“leptin resistance.” This resistance involves impaired
transport of leptin across the blood-brain barrier and
progressive inhibition of leptin signaling in the CNS, Nuclleus
particularly in the arcuate nucleus (, ). In mice, © 2018 Illustration Presentation ENDOCRINE SOCIETY

 hours of high-fat feeding is sufficient to reduce hy-


pothalamic responses to leptin (). Resistance has two Figure 2. The signaling form of the leptin receptor, also known as the long form or LepRb. Leptin
binds to the extracellular domain of LepRb, activating the associated Jak2 tyrosine kinase and
components: reduced access of peripheral leptin to its
promoting the Jak2-mediated phosphorylation of tyrosine residues 985, 1077, and 1138 of LepRb
sites of action in the hypothalamus and an intracellular (numbering is that of the murine receptor). Phosphorylated Tyr985 recruits PTPN11, which
signaling defect in leptin-responsive hypothalamic initiates the ERK signaling cascade. Phosphorylated Tyr1077 and Tyr1138 recruit STAT5 and STAT3,
neurons (). The extent to which resistance occurs in respectively, permitting their trafficking to the nucleus to mediate the control of cognate gene
the osteoblastic leptin receptors remains to be de- expression. In addition, mechanisms that limit LepRb signaling have been defined: Socs3, whose
termined. However, leptin resistance adds complexity expression is increased by leptin, binds to phosphorylated Tyr985 to blunt LepRb signaling. Also,
the tyrosine phosphatases, PTP1B and TCPTP, dephosphorylate Jak2 or LepRb to terminate LepRb
to the interpretation of leptin physiology. In a similar
signaling. In addition, Rho kinase 1, adenosine monophosphate activated protein kinase, insulin
manner to the obesity of leptin-deficient models receptor substrate (IRS)2, and SH2B1 have been shown to act downstream of LepRb, although the
being a reflection of inherent starvation responses that mechanisms by which LepRb controls these pathways have yet to be defined. Reproduced with
result when leptin signaling is reduced, the maintenance permission from Flak JN, Myers JMG. Minireview: CNS Mechanisms of Leptin Action. Mol Endocrinol
of obesity in long-term, high-fat-fed models similarly 2016; 30 (1): 3-12. [© 2018 Illustration Presentation ENDOCRINE SOCIETY].

doi: 10.1210/er.2017-00226 https://academic.oup.com/edrv 941


REVIEW

Figure 3. Signaling pathways by


which leptin interacts with the
hypothalamus and efferent pathways
acting from the hypothalamus to
the skeleton. [© 2018 Illustration
Presentation ENDOCRINE SOCIETY].

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


obesity, although absolute levels in the CNS were neuroendocrine abnormalities, such as hypogonadism.
higher in obese subjects than in nonobese. Therefore, The complete lack of leptin signaling is analogous to
in normal physiology (as distinct from pharmaco- extreme starvation and lipodystrophy, and as a re-
logical manipulations such as leptin injections into the sult the phenotypes of the ob/ob and db/db mice
CNS) leptin receptor occupancy is much higher in the are similarly extreme. The mice are infertile, have
periphery than in the CNS. These differences in cir- markedly reduced lean mass and brain weight, and
culating and CNS leptin concentrations are reflected in exhibit hypercortisolism, consistent with the stress of
the dose responses for the central and skeletal effects of near starvation.
this peptide defined in the elegant studies of Philbrick Ducy et al. () can be credited with bringing a
et al. (), which are discussed below. major focus on the influence of leptin on bone with
their description of the leptin-deficient ob/ob mouse.
This analysis focused on the trabecular bone of the
Bone Phenotypes of Animals With Reduced vertebral bodies, where a “high–bone mass phenotype”
Leptin Signaling was apparent. Trabecular bone volume was doubled in
the spine, but three-point bending of the femur
Reduced leptin signaling can be produced by the showed no difference from wild type, indicating that
absence of either a functional leptin gene or the gene the changes observed were site specific rather than
for its receptor. The ob/ob mouse is leptin deficient, generalized. The changes in trabecular bone volume of
and the db/db mouse and the fa/fa rat lack func- the vertebrae were unexpected because these mice
tional leptin receptors. As a result of the absence of a have high circulating glucocorticoid levels and are
leptin effect on eating behavior and energy metab- hypogonadal. The increase in vertebral bone mass
olism, these animals exhibit hyperphagia, early-onset appeared to be the result of increased bone formation,
severe obesity, insulin resistance, diabetes, and other approximately doubled, with no change in osteoblast

942 Reid et al Effects of Leptin on the Skeleton Endocrine Reviews, December 2018, 39(6):938–959
REVIEW

surface or number. Osteoclast numbers were increased and the close intercorrelation of fat mass and leptin
by #% in ob/ob mice, consistent with the doubling [r . . ()] means that multiple regression analysis
in deoxypyridinoline excretion observed. Osteoclast cannot be used to assess these interrelationships be-
numbers were returned to normal with sex hormone cause these variables are not sufficiently independent
replacement. These investigators observed essentially of one another. Failure to observe this caveat can result
the same phenotype in db/db mice, which are deficient in the finding of an inverse relationship between leptin
in the leptin receptor (Table ). and BMD (, ). One analysis concluded that leptin
There are now many other descriptions of the did not mediate the fat-BMD relationship, but it is not
skeletal phenotype of these animals (Table ), and it is clear that a statistical approach is able to determine the
clear that simply characterizing them as having a high direction of causation between biological variables,

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


bone mass does not adequately reflect the complexity particularly when leptin is so much more dynamic
of the skeletal response to leptin deficiency. Many than either BMD or fat mass (). The positive cor-
studies show that the long bones of the lower limb are relation between circulating leptin and bone mass has
shorter and have reduced BMD (, , –) (Fig. ). been documented across the lifespan: in neonates (),
Steppan et al. () assessed total body BMD and also children (), adolescents (), premenopausal women
found it to be reduced. Hamrick et al. (, ) and (), postmenopausal women (, , ), and men
Turner et al. () each studied vertebral and femoral (, ). Consistent with these bone density findings,
bone in the same animals. Both confirmed the Ducy leptin is reported to be lower in people with fractures
et al. () finding of increased trabecular bone volume (, ).The Health ABC assessed the relationship
in the spine (although Turner reported reduced bone between leptin and both prevalent and incident
formation rates), but both demonstrated a low–bone fractures in  older adults (). In that study,
mass phenotype in the femur. Hamrick et al. made the prevalent fractures were significantly less common
important observation that there is a dramatic increase with higher leptin levels (P = .) in women but not
in marrow adiposity in the femur of ob/ob mice, which in men. Incident fractures in women were also reduced
is not observed in the spine. by % and % in the middle and upper leptin tertiles,
In summary, the data in Table  indicate that leptin respectively, compared with the lower tertile (P ,
is critical to longitudinal skeletal growth and to the .), but these effects were lost after adjustment for
attainment of a normal BMD (–, , , , , BMI and race. As noted earlier, collinearity between
). Although trabecular bone in the spine is increased these variables is a problem in carrying out such
in most studies that measured it (, , ), the adjustments. Although these associations do not
suggestion that leptin has a uniformly negative effect establish causation, they suggest that, if anything, the
on the skeleton is not supported by these studies. effect of leptin on the human skeleton is a positive
one.

Associations of Circulating Leptin With Bone Bone turnover markers


Indices in Humans Many groups have examined relationships between
leptin and bone turnover markers. Some studies have
The phenotype of leptin-deficient animals or humans found no relationship (, ). Others report a positive
represents one end of what would be expected to be a relationship between circulating leptin and markers of
spectrum of leptin effects. Only small numbers of bone formation (, ) or an inverse relationship of
leptin-deficient humans have been assessed with re- leptin with bone resorption (, , ). Both the latter
spect to their skeletal phenotype, and it is important findings would suggest that leptin exerts a positive
to note that no consistent abnormalities emerged effect on bone mass, consistent with the studies of
(–). If leptin is involved in normal bone meta- BMD, but this effect is potentially confounded by the
bolism in humans, then variations in leptin levels other effects of fat mass.
within the normal range might be seen to be related to
various bone parameters. Such studies have been PTH and fibroblast growth factor-23
carried out for a variety of bone end points. PTH concentrations are directly related to adiposity in
most clinical contexts (, ), and circulating levels of
Bone density leptin and PTH are positively related to one another
There is now a substantial literature relating BMD or (). Body weight, fat mass, and leptin levels are higher
bone turnover to circulating leptin levels in cross- in patients with primary hyperparathyroidism (, ),
sectional studies in humans. Because both BMD and but leptin levels show no sustained change after cu-
leptin concentrations are related to fat mass, it would rative parathyroidectomy (, ), suggesting that
be expected that leptin and BMD would be positively PTH is not driving leptin secretion but that possibly
correlated, which is the finding in most studies the reverse may be the case. The latter suggestion is
(–). This association is sometimes found not to be supported by evidence that leptin increases circulating
independent of body weight or fat mass (, , ), PTH three to five times in ob/ob mice (, ). Both

doi: 10.1210/er.2017-00226 https://academic.oup.com/edrv 943


REVIEW

Table 1. Bone Phenotype of Animals With Reduced Leptin Signaling


Study Spine Appendicular

ob/ob Mouse

Ducy et al. 2000 (36) ↑ TBV, ↑↑ bone formation, ↑ osteoclasts Femur: normal strength

Steppan et al. 2000 (37) Femur: ↓ length and BMC

Total body: ↓ bone area, BMC and BMD

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


Hamrick et al. 2004 (38) ↑ Vertebral length, ↑ BMC, BMD and TBV, Femur: ↓ length, BMC, BMD, and cortical thickness; ↑↑ marrow adipocytes
↓ cortical thickness, marrow adipocytes normal

Hamrick et al. 2005 (39) ↑↑ Adipocytes, ↓ osteoclasts Total body: ↓ BMC and BMD

Tibia: ↓ bone formation

Kishida et al. 2005 (40) Growth plates: abnormal columnar structure, disorganized collagen fibrils,
↓ type X collagen, ↑ apoptosis, premature mineralization

Baldock et al. 2005 (41) Femur: ↑ TBV, trabecular thickness, mineral apposition rate,
osteoclast surface (all in cancellous bone)

Ealey et al. 2006 (42) Normal BMC, BMD, strength Femur: ↓ BMC, BMD, strength

Iwaniec et al. 2007 (43) ↑ Cancellous bone Femur: ↓ length, total bone volume; ↑ cancellous bone

Turner et al. 2013 (44) ↑ TBV, ↓ bone formation Femur: TBV normal, ↓ bone formation, ↓ linear growth; ↑ bone resorption

Philbrick et al. 2017 (35) Femur: ↓ linear growth, bone formation, trabecular thickness; bone resorption
normal

↓ Bone turnover markers (↓↓ CTX)

db/db Mouse

Lorentzon et al. 1986 (45) Tibia: ↓ length, BMC, BMD, cortical bone

Femur: ↓ TBV

Takeshita et al. 1995 (46) Femur: ↓ length, ash weight and BMD

Ducy et al. 2000 (36) ↑ TBV, ↑ bone formation, ↑ osteoclasts Femur: ↑ trabecular number (data not shown)

Ealey et al. 2006 (42) Normal BMC, BMD, strength Femur: ↓ BMC, BMD, strength

Williams et al. 2011 (47) ↓ Trabecular and cortical thickness Tibia: ↓ TBV, cortical bone
but not volume
Femur: ↓ strength and stiffness

↓ Serum osteocalcin

Turner et al. 2013 (44) ↓ Bone turnover markers

Spine: ↓ bone formation

Femur: ↓ bone formation

fa/fa Rat

Foldes et al. 1992 (48) Femur: shorter and lighter, ↑ cortical width

Mathey et al. 2002 (49) Femur: ↓ BMC, BMD, and strength

Tamasi et al. 2003 (50) Tibia: no change in BMD; ↓ TBV, osteoclast, and osteoid surface

Abbreviations: BMC, bone mineral content; CTX, C-telopeptide of type I collagen; TBV, trabecular bone volume.

leptin and its receptor are expressed in human direct and indirect effects of leptin on bone and of the
parathyroid chief cells, and leptin stimulates PTH leptin-independent effects of obesity on the skeleton,
release in vitro (). Any effects on bone turnover of accounting for the variable relationships between
leptin-driven PTH secretion will be modified by the leptin and turnover markers, described earlier.

944 Reid et al Effects of Leptin on the Skeleton Endocrine Reviews, December 2018, 39(6):938–959
REVIEW

Circulating levels of fibroblast growth factor- was decreased. They concluded that “ICV injection of
(FGF) are also correlated with adiposity (, ). leptin promotes expression of pro-osteogenic factors
Leptin directly stimulates FGF expression in pri- in bone marrow, leading to enhanced bone formation
mary rat osteoblasts (), and administration of leptin in ob/ob mice” (). The study by Pogoda et al. () is
to ob/ob mice almost doubles serum FGF concen- particularly important because it extends this work
trations and is associated with reduced renal expres- beyond rodent studies. Using adult ewes infused with
sion of a-hydroxylase mRNA and reductions in leptin ICV for  months at a dosage that did not
circulating ,-hydroxyvitamin D (, ). reduce body weight, they showed a global reduction in
bone formation, as reflected by circulating bone al-
kaline phosphatase and osteocalcin concentrations,

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


Skeletal Effects of Leptin Action in the CNS and a reduction in bone formation on histomor-
phometry. Trabecular bone volume was reduced in
After the identification of leptin as a key regulator of the lumbar spine, iliac crest, and distal femur, but
energy homeostasis, work proceeded rapidly to elu- mechanical testing of the tibia was unaffected. Thus,
cidate its mechanism of action. This research identified Table  demonstrates the consistency of evidence that
the CNS, particularly the hypothalamus, as the key site ICV leptin reduces trabecular bone volume in the
of action of leptin. Accordingly, some early work to spine and elsewhere, but it is not clear that this is
determine the actions of leptin on the skeleton focused always caused by reduced bone formation, and effects
on administration of leptin into the CNS. These on cortical bone are mixed.
studies are summarized in Table . The Karsenty group () went on to demonstrate
The first such study was carried out by Ducy et al. that the bone and anorexigenic effects of central leptin
(), who administered intracerebroventricular (ICV) treatment were dissociable. In wild-type mice, the
leptin infusions to ob/ob and wild-type mice. There selective destruction of neurons in the ventral hypo-
was no detectable leak of leptin into the bloodstream. thalamus by gold thio-glucose increased vertebral
Trabecular bone volume of the vertebrae was sub- cancellous bone mass, and ICV leptin treatment ( ng/h)
stantially reduced in both groups to below wild-type in these animals no longer influenced the cancellous
levels by the leptin treatment. Weight change was not phenotype, whereas it decreased body weight by %.
reported. Bone formation was increased in the ob/ob Neuropeptides mediating leptin’s anorexigenic function
mouse before treatment (see Table ), but changes in (a-melanocyte-stimulating hormone, CART) did not
bone formation were not reported after ICV leptin affect bone formation. In parabiosed ob/ob mice, ICV
treatment, although the authors concluded that “leptin leptin corrected the skeletal changes in the recipient, with
is a major regulator of bone formation.” Similar
findings in the vertebrae were reported by Iwaniec
et al. (), who gave a single hypothalamic ICV in-
jection of leptin-producing adenoassociated virus to
ob/ob mice, which resulted in normalization of body
weight and recapitulation of the wild-type skeletal
phenotype. Femoral and vertebral cancellous bone
volume were reduced, but femur length and total
femur bone volume were increased to wild-type levels,
reinforcing the antiosteogenic effect of ICV leptin on
cancellous bone but demonstrating anabolic actions
on cortical bone. Turner et al. () also reported the
effect of ICV injection of a leptin-producing construct
on vertebral bone. This treatment restored bone
microarchitecture to normal but was accompanied
by a fourfold increase in osteoblast perimeter in the
spine, indicating that it is an oversimplification to
characterize hypothalamic leptin as simply being
“antiosteogenic” in its skeletal effect. Bartell et al. ()
also showed that ICV leptin increased mineral ap- Figure 4. Contrasting bone phenotypes of the ob/ob mouse
position rate in both the vertebra and the tibia, are demonstrated by the reduced length of the femur in the
comparably to the effect of subcutaneously adminis- ob/ob but an increase in trabecular bone volume, shown in the
insets. Body weight is approximately doubled in the ob/ob.
tered leptin. They found that expression of genes
Reproduced with permission from Iwaniec UT, Boghossian S,
associated with osteogenesis was increased in femoral Lapke PD, et al. Central leptin gene therapy corrects skeletal
bone marrow stromal cells after ICV injection; abnormalities in leptin-deficient ob/ob mice. Peptides 2007;
whereas expression of genes associated with osteo- 28:0.1012-1019. [© 2018 Illustration Presentation ENDOCRINE
clastogenesis, adipogenesis, and adipocyte lipid storage SOCIETY].

doi: 10.1210/er.2017-00226 https://academic.oup.com/edrv 945


REVIEW

Table 2. Effects of ICV Leptin Administration on Bone in Animals


Study Model Dosing Effects

Leptin-deficient

Ducy et al. (2000) (36) 4-mo-old ovariectomized ob/ob 8 ng/h over 28 d Spine TBV ↓ from 16.9% to 8.4%
mice
Weight changes not reported

Iwaniec et al. (2007) (43) 8- to 10-wk-old male ob/ob mice Single hypothalamic ICV Body weight ↓ 50%
injection of leptin-producing

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


adenoassociated virus ↑ Femoral length and total bone volume, ↓ femoral and
vertebral cancellous bone volume 15 wk postinjection

ob/ob mice no longer differed from WT

Bartell et al. (2011) (84) 15-wk-old female ob/ob mice 0.4 or 1.5 mg/d for 12 d Body weight ↓, whole body BMD ↑

↑ Osteogenesis in femoral bone marrow stromal cells

↓ Serum insulin and ↑ osteocalcin

Turner et al. (2013) (44) 8- to 10-wk-old male ob/ob mice Single hypothalamic ICV Body weight ↓ 50%
injection of leptin-producing
adenoassociated virus Spine: TBV ↓ 50%, osteoblast perimeter ↑ 43

Wild-type

Ducy et al. (2000) (36) 4-mo-old female mice 8 ng/h over 28 d TBV ↓ from 13.2% to 9.4%

Weight changes not reported

Pogoda et al. (2006) (85) 6- to 7-y-old ewes 8 mg/h for 3 mo Body weight: no change

Tibia: no change in mechanical testing

TBV ↓ in lumbar spine, iliac crest, and distal femur

↓↓ Bone formation rate, bone alkaline phosphatase, and


osteocalcin

Abbreviations: TBV, trabecular bone volume; WT, wild type.

no change in the parabiosed animal, ruling out a cir- treatment with a b-agonist decreased bone mass and
culatory factor in the bone effects of central leptin and bone formation rate in the tibial and vertebral meta-
suggesting a neuronal mediator. physes and that a b-blocker (propranolol) had the op-
posite effect in wild-type mice. Leptin ICV infusion
Mediation of leptin effects on bone via the SNS decreased fat pad weight but not bone mass in b-blocked
Regulation of the SNS by the hypothalamus, and the wild-type mice. They also found that propranolol could
known effects of adrenergic agonists on bone, sug- completely prevent bone loss after ovariectomy in wild-
gested the SNS as a promising candidate for mediating type mice.
the central effects of leptin. It was already established Subsequent studies from the Karsenty group ()
that leptin increases catecholamine secretion, an effect provided further evidence of leptin acting on bone
thought to be mediated by the ventromedial hypo- formation via the SNS and extended this evidence
thalamus (). Takeda et al. () examined a model further to suggest that this pathway also regulated
that impairs the SNS by blocking the production bone resorption. Mice lacking the b-adrenergic
of norepinephrine and epinephrine (dopamine b- receptor (Adrb2/2) had increased trabecular bone
hydrolase deficient mice, Dbh2/2) and reported a similar volume in both the vertebrae and the distal femur.
but less severe vertebral cancellous bone phenotype to Transplantation of wild-type bone marrow cells
that of the ob/ob mice. Cortical bone was not assessed. into irradiated Adrb2/2 mice reduced bone for-
Cancellous bone in the Dbh2/2 mice was not affected by mation to normal, and, conversely, transplantation of
central leptin treatment, despite this intervention dra- Adrb2/2 bone marrow cells into irradiated wild-
matically reducing the weight of the gonadal fat pad. type mice significantly increased bone formation.
Thus central leptin could act on bone via the SNS in- ICV leptin did not affect vertebral cancellous bone in
dependently of the pathways that mediate its effects on Adrb2/2 mice, confirming the role of the SNS in
body weight. They then demonstrated that systemic this response.

946 Reid et al Effects of Leptin on the Skeleton Endocrine Reviews, December 2018, 39(6):938–959
REVIEW

Bone resorption was also found to be altered by Rejnmark et al. () found comparable benefit to that
the leptin-SNS signaling pathway, via central and of b-blockers with ACE inhibitors and calcium
peripheral actions (). Leptin deficiency reduces channel blockers (suggesting that this effect is related
expression of the neuropeptide CART in the hypo- to hypertension or its treatment rather than b-
thalamus, promoting bone resorption. In bone, local blockers per se); and de Vries et al. () found that the
b-adrenergic receptor signaling increases RANKL protective effect of b-blockers was present only among
expression from osteoblast progenitor cells, with patients with a history of use of other antihypertensive
RANKL expression lower in Adrb2/2 osteoblasts (, agents and was not associated with cumulative dose or
), whereas the b-adrenergic agonist isoproterenol receptor selectivity of the b-blockers. Thus, it is likely
increases osteoblast-induced osteoclastogenesis from that observational studies of the use of b-blockers are

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


bone marrow monocytes. Thus, leptin signaling from confounded by the indications for which these drugs
the brain via the SNS acts on osteoblasts to influence are prescribed and by other medications that are
not only bone formation but also RANKL expression commonly coprescribed with them. Also, although
and subsequently osteoclast differentiation and can- early agents in this class (e.g., propranolol) were
cellous bone resorption. These findings were rein- nonselective, b-selective blockers are now more
forced by a subsequent article from this group commonly used and will not affect osteoblasts that
demonstrating that cell-specific deletion of the leptin express only the b receptor.
receptor in neurons resulted in increases in bone Randomized studies can overcome these potential
formation, increases in bone resorption in the verte- confounders in observational data. The many trials
brae, and increased trabecular volume in the spine and involved in the development of b-blockers as clinical
distal tibia (). Additionally, the l/l mice, which have a drugs have not reported reduced fracture numbers,
gain-of-function mutation in the leptin receptor, although these data would have been captured as
showed upregulation of sympathetic activity and re- adverse events in these studies. To address this
duced spine and tibia trabecular bone volume, al- question more formally, we analyzed data pooled from
though gonadal fat pads were reduced (). nine randomized controlled trials of a nonselective
The SNS also mediates leptin’s regulation of lipid b-blocker (carvedilol) in the management of con-
metabolism. Acting through the hypothalamus and gestive heart failure (). This patient population was
the SNS, leptin inhibits lipogenesis and promotes li- chosen because it was likely to have a high risk of
polysis and fatty acid oxidation in both adipose tissue fracture. From a total subject population of , .%
and the liver (, ). These effects also occur in bone of placebo-treated subjects experienced a fracture
marrow adipocytes: direct injection of leptin into the compared with .% of those randomized to carvedilol
ventromedial hypothalamus in rats substantially de- (relative risk, .; % CI, . to .). This database
creases the adipocyte population in bone marrow, does not provide any evidence to support the hy-
primarily through apoptosis (). Thus, the central pothesis that b-blockers reduce fracture numbers. We
actions of leptin are likely to directly affect local bone also carried out a prospective randomized controlled
cell activity as a result of changes in adipokine and fatty trial of propranolol in normal postmenopausal women
acid production in marrow adipocytes (). to assess its effects on bone turnover. This study did
not demonstrate stimulation of bone formation, as had
Effects of adrenergic blockade on bone in humans been observed in mice, but found variable declines in
As noted earlier, the bone loss in leptin-deficient and markers of bone turnover, particularly serum osteo-
wild-type mice receiving infusions of leptin into the calcin (). Thus, the human studies do not re-
third ventricle results from inhibition of bone for- capitulate the findings in mice, although we do not
mation () and stimulation of bone resorption (). have comprehensive dose-response data for selective
Blockade of the SNS abrogates these effects, which and nonselective b-blockers to exclude the possibility
appear to be mediated by b-adrenoreceptors on that these agents can affect skeletal mass and fractures.
osteoblasts (). Drugs that modify the action of the In light of currently available information, the SNS
SNS have been widely used clinically for many years, so does not seem to be a clinically important regulator of
if sympathetic activity has a measurable impact on bone metabolism in leptin-sufficient humans.
bones in humans, this effect should be evident from
epidemiological studies and clinical trials. Many Leptin and NPY
groups have undertaken such analyses, demonstrating As mentioned earlier, leptin’s metabolic actions are
beneficial (–), null (, ), or detrimental () mediated in part through regulation of energy
effects of b-blockers on fractures. Among the studies modulatory neuropeptides in the hypothalamus, in-
suggesting apparent benefit, it should be noted that cluding pro-opiomelanocortin, CART, agouti-related
Yang et al. () found benefit only in those using peptide, and NPY. Along with CART, NPY has been
selective b-blockers, which should not affect ad- shown to mediate important aspects of leptin’s action
renergic signaling in osteoblasts (suggesting that this on bone, with increased expression of NPY in the
study should be classified as null in the list above); hypothalamus in ob/ob mice (). NPY has a strong

doi: 10.1210/er.2017-00226 https://academic.oup.com/edrv 947


REVIEW

antianabolic influence on cortical and cancellous bone, mediated by a pathway from the brainstem to the
and NPY-deficient mice display increases in cancellous arcuate nucleus, via Htra and b serotonin receptors
and cortical mass (). Consistent with this action, (). Therefore, centrally leptin appears to exert an
dual NPY- and leptin-deficient (NPY2/2ob/ob) mice indirect osteogenic action through the suppression of
display greater whole body, femoral, and vertebral serotonergic signaling to the ventromedial hypothal-
BMD compared with ob/ob mice (), the result of an amus, but efferent pathways are needed for this signal
increased cortical bone formation rate correcting the to be relayed to bone cells. Figure  describes these
reductions in BMD of ob/ob mice to wild-type levels, pathways.
with minimal effects on body composition ().
Consistent with the segregation of cancellous and

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


cortical control in ob/ob mice, no changes in cancel- Direct Actions of Leptin on Bone
lous bone were apparent between NPY2/2ob/ob and
ob/ob mice, suggesting that the leptin-NPY pathway to Receptors
cortical bone can function independently of the leptin- In addition to the CNS-mediated effects of leptin on
adrenergic axis to cancellous bone. Thus the reduction bone, discussed earlier, it has become firmly estab-
in cortical bone mass evident in ob/ob mice might be lished that leptin also acts directly on bone cells. The
contributed to by an elevation in central NPY ex- signaling form of the leptin receptor has been dem-
pression, whereas reduction of adrenergic signaling onstrated in rat osteoblasts (, ); human oste-
produces the increase in cancellous bone volume. oblasts (, ); human mesenchymal stem cells
Importantly, this complex, envelope-specific regula- undergoing osteogenic () or adipogenic differenti-
tion of bone turnover by leptin deficiency is consistent ation (); human (), porcine (), and mouse
with the skeletal response to calorie deprivation evi- () chondrocytes; and osteoblastic and chondrocytic
dent in wild-type mice (). Consistent with cell lines (, ). There is some evidence that leptin
increased central NPY mimicking a low-leptin/ receptor expression decreases as osteoblast differen-
starvation response, viral-mediated overexpression tiation advances (). In contrast, Ducy et al. ()
of NPY in the hypothalamus led to marked increases reported absence of leptin receptors in primary os-
in food intake and in gains white adipose tissue mass; teoblasts and whole bone. However, the balance of
however, despite this increase in weight, cortical bone these results indicates that leptin has the potential to
formation was markedly reduced (). Hypothalamic play a direct role in skeletal physiology, independent of
NPY signaling is an important part of the central the CNS. This potential is confirmed by in vitro studies
regulation of bone by leptin, most prominently in in skeletal cell and tissue cultures showing that leptin
cortical bone, reducing bone mass in leptin-deficient activates multiple pathways, including Jak/Stat, in
states. Thus the ob/ob mouse may be an accurate cultured osteoblast lineage cells (–).
representation of the skeletal response to reduced
leptin signaling, implicating both adrenergic and NPY- Osteoblasts
mediated pathways in regulating hypocaloric re- A consistent body of evidence shows that leptin has a
sponses in bone. direct anabolic effect on cells of the osteoblast lineage.
Iwaniec et al. () reported dose-related increases in
Leptin and serotonin the formation of mineralized bone nodules in primary
Serotonin is a monoamine neurotransmitter produced osteoblast cultures with leptin in concentrations be-
in the CNS by tryptophan hydroxylase  (Tph) and in tween  and  ng/mL. These findings were sub-
the gastrointestinal tract by Tph (). Because se- sequently confirmed by Reseland et al. (). Thomas
rotonin does not cross the blood-brain barrier, central et al. () showed that leptin promoted differentiation
serotonergic actions are not influenced by serum se- of a human marrow stromal cell line into an osteoblast
rotonin levels. Tph-expressing neurons express the phenotype, increasing synthesis of the bone matrix
leptin receptor, and leptin reduces Tph expression proteins type I collagen and osteocalcin. Differentia-
and activity in serotonergic neurons (). Tph2/2 tion of these cells to adipocytes was diminished by
mice display a low bone mass phenotype resulting leptin (). Cornish et al. () demonstrated stim-
from reduced bone formation and increased re- ulation of osteoblast proliferation by leptin at physi-
sorption. Ablation of the leptin receptor on seroto- ological concentrations. The maximum stimulation of
nergic neurons produced a high cancellous bone mass cell growth was comparable to that produced by other
phenotype similar to ob/ob. The phenotypic similarity major osteoblast mitogens, such as IGF, TGF-b,
suggested a functional link between brainstem neu- epidermal growth factor, and insulin (Fig. ). In
rons and the ventromedial hypothalamus in the cultures of human iliac crest osteoblasts, Gordeladze
central control of bone mass, reinforced by a high et al. () showed that leptin promoted collagen
cancellous bone mass phenotype after deletion of the synthesis, cell differentiation, and in vitro minerali-
serotonin receptor (Htrc2/2) from the ventromedial zation, as well as cell survival and transition into
hypothalamus in mice. The regulation of appetite is preosteocytes. Gene expression changes with leptin

948 Reid et al Effects of Leptin on the Skeleton Endocrine Reviews, December 2018, 39(6):938–959
REVIEW

treatment indicate increased osteoblast differentiation number and thymidine incorporation into canine
and mineralization (). An analysis of the human and ovine chondrocytes after treatment with phys-
osteoblast transcriptome concluded that these cells iological concentrations of leptin. They also showed
expressed both leptin and its receptor, that leptin an increase in growth plate thickness after systemic
expression was substantially upregulated by gluco- administration of leptin to normal adult male mice.
corticoid, and that leptin might be an important Maor et al. () confirmed the presence of leptin
autocrine regulator in bone (). In vivo, leptin in- receptors in chondrocytes and demonstrated in or-
fusions increase mRNA levels for bone and cartilage gan cultures of mandibular condyles that leptin in-
matrix proteins in tibiae of ob/ob mice (). Leptin also creased the width of the chondroprogenitor zone,
appears to play a role in fracture healing. In ob/ob inducing both proliferation and differentiation of

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


mice, one group has reported that maturation of chondrocytes. Leptin also increased the abundance
fracture callus is delayed, and this effect is fully re- of the IGF receptor within the chondrocyte and
versed with local application of leptin (); another the progenitor cell populations, suggesting that it
study has found that ob/ob mice show accelerated acts synergistically with other growth factors. Gat-
callus formation and remodeling but that the cortex Yablonski et al. () found that leptin increased the
formed is thinner (). level of PTH-related peptide and reduced that of
Turner et al. () used an innovative approach to Indian hedgehog protein in murine growth plates,
establish the local effects of leptin on bone formation. using both in vivo and in vitro models. They con-
Wild-type mice were lethally irradiated, and then their cluded that leptin plays a key role in regulating
bone marrow was reconstituted with marrow from chondrocyte proliferation and differentiation in the
either wild-type or db/db mice. Engraftment of db/db growth plate via these mechanisms. Leptin has also
bone marrow did not influence energy homeostasis, been shown to increase muscle mass in aged mice
but it reproduced the low rates of bone formation ob- ().
served in db/db mice. These results indicate that oste-
oblast behavior in vivo is determined by the presence or
absence of the leptin receptor in bone and that the bone Systemic Administration of Leptin
phenotype of the db/db mouse can be recapitulated by
the absence of leptin signaling in bone cells alone. The principal studies assessing the effects on bone of
the systemic leptin administration are set out in
Osteoclasts Table . Studies have been carried out in leptin-
The first suggestion that leptin might locally regulate deficient mice and in normal animals.
bone resorption came from Holloway et al. (),
who demonstrated leptin inhibition of osteoclast Leptin-deficient animals
generation in cultures of human peripheral blood In  Liu et al. () showed that leptin replacement
mononuclear cells. Leptin increased osteoprotegerin in ob/ob mice increased osteoblast number .-fold,
mRNA and protein expression in cultures, suggesting
that the inhibitory effect was mediated via the
RANK/osteoprotegerin system. Burguera et al. ()
found that leptin increased osteoprotegerin (a pro-
tein that inhibits osteoclastogenesis) and decreased
RANK ligand levels (the principal regulator of os-
teoclast development) in human marrow stromal
cells. Lamghari et al. () found that leptin reduced
RANKL mRNA and protein in the MCT-E cell
line, and Gordeladze et al. () showed leptin
stimulation of osteoprotegerin in osteoblasts, en-
tirely consistent with the Holloway results. Work by
Cornish et al. () complemented these findings. They
reported reduced osteoclast formation in mouse bone Figure 5. Response of thymidine incorporation in primary
marrow cultures treated with leptin but no effect of leptin cultures of fetal rat osteoblasts at 24 h to a maximal dose of
on the activity of mature osteoclasts, either those isolated leptin, compared with responses to maximal concentrations
from rats or in neonatal organ culture. Thus, leptin of a number of other osteoblast growth factors. Data are
appears to act on the osteoblast to regulate osteoclas- treatment/control ratios and presented as mean 6 SEM. P ,
0.01. *, significantly different from control; EGF, epidermal
togenesis but not on the mature bone resorbing cell.
growth factor. Reproduced with permission from Cornish J,
Callon KE, Bava U, et al. Leptin directly regulates bone cell
Other skeletal cells function in vitro and reduces bone fragility in vivo. J Endocrinol
As noted earlier, chondrocytes have leptin receptors. 2002; 175 (2):405-415. [© 2018 Illustration Presentation
Cornish et al. () demonstrated increases in cell ENDOCRINE SOCIETY].

doi: 10.1210/er.2017-00226 https://academic.oup.com/edrv 949


REVIEW

Table 3. Effects of Systemic Leptin Administration on Bone in Animals


Study Model Dosing Effects

Leptin-deficient

Liu et al. 1997 (130) ob/ob female mice Leptin 50 mg/d bid IP for 2 or 4 wk Body weight ↓ 46%, IGF1 ↑ 40%

Tibia: all indices of bone formation ↑↑, no change


in osteoclast numbers

Steppan et al. 2000 (37) 4-wk-old ob/ob male mice Leptin 50 mg/d IP for 3 wk Femur: ↑ length and BMC

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


Total body: ↑ bone area, BMC, and BMD

Hamrick et al. 2005 (39) 15-wk-old ob/ob female mice Leptin 2.5 or 10 mg/d SC for 2 wk Fat mass ↓ 43%

Spine: ↓ adipocytes, ↑ osteoclasts

Total body: ↑ BMC and BMD

Femur: ↓ adipocytes

Tibia: ↑ bone formation

Kishida et al. 2005 (40) 4-wk-old ob/ob mice Leptin 50 mg/d IP for 2 wk ↑ Femoral and humeral lengths, ↓ premature
mineralization of growth plates

Bartell et al. 2011 (84) 15-wk-old ob/ob female mice Leptin 10 mg/d SC for 12 d ↓ Weight

↑ Whole body BMD, ↑ muscle mass

Spine: ↑ mineral apposition rate, ↓ marrow


adipocytes

Tibia: ↑ mineral apposition rate, ↓ marrow


adipocytes

↓ Serum insulin; ↑ serum IGF1, OPG, osteocalcin,


pyridinoline, RANKL

Turner et al. 2013 (44) 12-wk-old female ob/ob mice Leptin 40 mg/d SC for 3 wk ↓ Weight

Spine: ↓ TBV, ↑ bone formation

Femur: ↑ bone formation, ↑ linear growth, ↓


osteoclasts

Philbrick et al. 2017 (35) 6-wk-old female ob/ob mice Leptin infusion (0, 4, 12, 40, 140, or ↑ Bone formation and gene expression (bone and
400 ng/h) for 12 d via SC osmotic cartilage matrix proteins, bone morphogenetic
pumps protein signaling) in tibia at infusion rates not
changing energy metabolism or hypothalamic
gene expression

Wild-type

Steppan et al. 2000 (37) 4-wk-old wild-type male mice Leptin 50 mg/d IP for 3 wk No significant effects in femur or total body

Burguera et al. 2001 (125) Ovariectomized rats Leptin 100 mg/d SC for 1 mo Tibia: ↑ TBV and trabecular number, ↓ bone
formation rate

Cornish et al. 2002 (109) Normal male mice Leptin 43 mg/d SC for 4 wk Tibia: ↑ growth plate thickness, ↓ fragility, no
change in bone histomorphometry

Gat-Yablonski et al. 2004 (131) Prepubertal male mice Leptin 8 mg/g body weight per ↓ Weight gain
day IP
Tibia: ↑ length compared with pair-fed controls;
↑ growth plate width, chondrocytes, and
IGF-IR

Hamrick et al. 2005 (39) 15-wk-old female mice Leptin 2.5 or 10 mg/d SC for 2 wk Total body: No change BMD and BMC
(Continued )

950 Reid et al Effects of Leptin on the Skeleton Endocrine Reviews, December 2018, 39(6):938–959
REVIEW

Table 3. Continued
Study Model Dosing Effects

Martin et al. 2005 (132) Tail-suspended and control Leptin 0.35 mg/kg/d IP for 3–14 d Tibia: ↓ bone loss from tail suspension, ↑ bone
female rats formation, ↓ bone resorption, ↓ OPG in bone,
↓ adipocytes in marrow

Martin et al. 2007 (133) Tail-suspended and control Leptin 50 and 500 mg/kg/d IP for High dose ↓ body weight
female rats 2 wk
Femur: ↑ BMD with low dose, ↓ BMD with high

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


dose

Tibia: ↑ BMD with low dose, ↓ BMD and bone


formation with high dose; leptin ↓ adipocyte
volume in tibial medulla

↑ CTX, ↓ osteocalcin

Baek et al. 2009 (134) Tail-suspended and control Leptin analog (LY35510), 0.35 mg/ Tibia: ↑ BMD, ↑ mineral apposition rate, and ↓
male rats kg/d SC resorbing surface in tail-suspended rats

Bertoni et al. 2009 (135) Pregnant mice Leptin 2 mg/kg SC on gestation Longer and wider ossification centers in neonates
days 7, 9, and 11 after leptin

Stunes et al. 2012 (136) Female rats Leptin 100 or 200 mg/d SC for 9 wk ↓ Weight

Femur: no change in BMD; in 100 mg/d group ↓


cortical volume and strength

↓ Whole body BMD in 100 mg/d group

Abbreviations: BMC, bone mineral content; CTX, C-telopeptide of type I collagen; OPG, osteoprotegerin; SC, subcutaneous; TBV, trabecular bone volume.

with accompanying substantial increases in bone are unchanged or decreased (Table ). In contrast to
formation rates. The nonskeletal phenotype of the ob/ the appendicular findings, in the spine, where tra-
ob mouse is also rapidly reversible with the systemic becular bone volume is increased in untreated ob/ob
administration of leptin. After  weeks leptin ad- mice, this abnormality was reversed by leptin ad-
ministration to ob/ob mice, Steppan et al. () ministration, associated with an increase in osteoclast
demonstrated a % decrease in food intake and a % numbers (, ). Hamrick et al. () showed that the
decrease in total body fat, resulting in a % decrease expansion of adipocyte mass in both the femur and
in body weight. Liu et al. found a % decrease in body spine is reversed by leptin treatment.
weight. Some studies have not reported these nutri- The study by Philbrick et al. () is particularly
tional changes during leptin treatment, but these ef- interesting in that it provides details of the dose-
fects are substantial and likely to contribute to the response for the skeletal and nonskeletal effects of
bone changes that occur, particularly with central leptin in ob/ob mice. Half-maximal effects on energy
leptin administration where near-starvation can result metabolism required leptin dose rates of  to  ng/h,
from its anorectic effects. Weight loss in normal mice whereas  to  ng/h produced half-maximal effects
significantly reduces cortical bone mass (), and on linear growth and on histomorphometric and
femoral and vertebral bone mass is closely related to biochemical indices of bone formation (Fig. ).
body mass at baseline (r = . to .) and after a Changes in histomorphometric indices of osteoblast
high-fat diet to the increase in body mass in mice activity were seen at the lowest dosage studied ( ng/h),
(). The near-normalization of the bone phenotype and longitudinal growth was increased at  ng/h, even
over a -week period in the Steppan study (see Table ) though these infusion rates achieved circulating leptin
in the face of what would usually be regarded as concentrations ,% of those in wild-type animals.
adverse changes in soft tissue mass strongly suggests an Blood glucose, abdominal fat mass, and uterine weight
anabolic effect of leptin on bone (). were unaffected by these infusion rates, although these
Across all the studies of leptin administration in ob/ parameters were affected at infusion rates of  and
ob mice, there is a consistent finding of reduced body  ng/h, which achieved physiological and supra-
weight but increases in appendicular bone formation, physiological circulating leptin levels, respectively.
linear bone growth, and bone density after leptin Interestingly, the marked suppression of serum
administration. Although leptin produces substantial C-telopeptide of type I collagen (CTX, a specific
positive effects on bone formation, there is in- measure of bone resorption) in the untreated ob/ob
consistency in the effects on osteoclast numbers, which mouse was reversed only by these much higher

doi: 10.1210/er.2017-00226 https://academic.oup.com/edrv 951


REVIEW

infusion rates, suggesting that leptin effects on bone higher. This higher dosage reduced fat mass by %
resorption are mediated by a different pathway from over  weeks, which might explain the negative bone
that regulating formation. effects of this pharmacological dosage. Because leptin
reduces body weight, and this itself is likely to affect
Wild-type animals bone mass, Gat-Yablonski did studies in which ani-
The results of leptin treatment in wild-type animals are mals were pair-fed, to disentangle the direct effects of
less consistent. Steppan et al. () and Hamrick et al. leptin on the bone from those mediated by weight loss
() found no changes in bone density with leptin (). These studies made clear that weight loss
treatment and no effects on histomorphometry, al- negatively affects linear growth and growth plate
though skeletal fragility assessed with three-point thickness, but that this is reversed by leptin treatment.

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


bending of the tibia was diminished in the Cornish In summary, the effects of leptin administration were
et al. study (). Burguera et al. () found that leptin less dramatic in wild-type animals than in leptin de-
reduced postovariectomy bone loss and inhibited the ficiency, but leptin remained anabolic to bone unless
increase in bone turnover associated with ovariectomy, given in very high doses resulting in marked anorexia
similar to the effect of estrogen. Several studies have and weight loss.
found positive effects on the growth plate or on linear
growth, and some reported reduced bone resorption Human studies
(, ). Martin et al. () demonstrated beneficial The literature describing leptin treatment of humans is
effects from leptin  mg/kg/d but negative effects on small and is summarized in Table . All studies used
BMD in the femur and tibia with a dose  times recombinant human methionyl leptin administered

Figure 6. Dose-response comparison of the effects of 12-d subcutaneous leptin infusions on bone formation rate in the distal femur
(upper left panel), Ucp1 gene expression in brown adipose tissue (BAT) (upper right panel), and on bone resorption measured as serum
C-telopeptide of type I collagen (CTX) in ob/ob mice. The bone formation effects are likely to reflect the direct effects of leptin on
osteoblasts, and the actions on Ucp1 reflect leptin’s effects on energy metabolism, probably mediated through the CNS. Unlike the
various measures of bone formation assessed, serum CTX (lower panel) was normalized only at high leptin infusion rates, suggesting an
indirect mechanism for this action. The x-axes show leptin infusion rates throughout the 12-d experimental period. The greater
sensitivity to leptin of bone compared with energy metabolism was reflected in activation of Jak/Stat signaling in the tibia at much
lower leptin infusion rates than were needed for activation of Jak/Stat signaling in the hypothalamus (data not shown). Data are
mean 6 SEM. “a” indicates different from vehicle-treated ob/ob mice (ob/ob 0), P , 0.05. Wild-type (WT) mice are shown as
a reference group. Reproduced with permission from Philbrick KA, Wong CP, Branscum AJ, et al. Leptin stimulates bone formation in
ob/ob mice at doses having minimal impact on energy metabolism. J Endocrinol 2017; 232 (3):461-474. [© 2018 Illustration
Presentation ENDOCRINE SOCIETY].

952 Reid et al Effects of Leptin on the Skeleton Endocrine Reviews, December 2018, 39(6):938–959
REVIEW

Table 4. Effects of Leptin Administration on Bone in Humans


Study Patients Regimen Bone Effects Other Effects

Farooqi et al. 1999 (54) 9-y-old girl with Met-leptin 0.028 mg/kg Total body BMC ↑ 0.15 Weight 94.4 kg → 78.0
congenital leptin of lean mass per day kg kg; food intake ↓ 40%
deficiency for 12 mo

Simha et al. 2002 (51) 2 women with Met-leptin 0.02–0.08 No change in BMD Weight ↓ 2–3 kg
generalized mg/kg/d for 16–18 (spine, hip, total body,
lipodystrophy mo radius); no change in

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


bone turnover
markers or PTH

Moran et al. 2004 (139) 3 men and 11 women Met-leptin 0.01–0.06 No change in BMD Weight ↓ 3.3 kg
with generalized (12) mg/kg/d for 12 mo (total body); no
or partial change in bone
lipodystrophy turnover markers

Welt et al. 2004 (140) 8 women with Met-leptin 0.08 mg/kg/ ↑ Bone alkaline Weight ↓ 2.5 kg; ↑ LH,
hypothalamic d for 3 mo phosphatase and estradiol, thyroid
amenorrhea osteocalcin hormones, IGF1, IGF-
binding protein-3

Chou et al. 2011 (141) 20 women with Met-leptin 0.08 mg/kg/ ↑ Osteocalcin and Fat mass ↓ 2 kg despite
hypothalamic d for 36 wk with transient ↑ urine NTX leptin dosage
amenorrhea dosage adjustment reductions; return of
randomly assigned, 7 menstruation in 70%
on leptin completed and normalization of
gonadal, thyroid,
growth hormone,
and adrenal axes

Conroy et al. 2011 (142) 8 women, 4 men (2 Met-leptin 0.08 mg/kg No change in PINP, Weight stable at 10%
nonobese, 10 obese) fat mass per day in BSAP, NTX, or PTH below baseline
being maintained at men and 0.14 mg/kg
baseline weight fat mass per day in
210% women, with dosage
adjustment

Sienkiewicz 2011 (143) 6 women with Met-leptin 0.04–0.012 ↑ Spine BMD, ↓ CTX Weight ↓ 3%; ↑ IGF1
hypothalamic mg/kg/d for another
amenorrhea from 6–12 mo
Chou study

Foo et al. 2014 (144) 18 women with Met-leptin 0.08 mg/kg/ ↓ PTH at 36 wk and in Weight changes not
hypothalamic d for 36 wk with RANKL/OPG ratio reported
amenorrhea dosage adjustment
randomly assigned,
10 to leptin
Met-leptin was given subcutaneously in all studies.
Abbreviations: BMC, bone mineral content; BSAP, bone-specific alkaline phosphatase; NTX, N-telopeptide of type I collagen; OPG, osteoprotegerin; PINP, procollagen type 1 N-terminal
propeptide.

subcutaneously once or twice a day. The Farooqi study ) or a small increase (). Bone turnover markers
et al. () is a case report of a person with congenital were either unchanged (, , ) or increased (,
leptin deficiency who had a % decrease in food ), apart from a reduction in the bone resorption
intake accompanied by profound weight loss during a marker CTX in the Sienkiewicz studies ().
year of leptin treatment. These nutritional changes Whereas Simha and Conroy both reported no
were associated with a moderate increase in total body change in parathyroid hormone concentrations, Foo
bone mineral content. In contrast, the cases of lipo- found that PTH levels were reduced by %, although
dystrophy or hypothalamic amenorrhea had low body this change became apparent only between  and
weights at baseline, but showed further loss of fat  weeks of leptin treatment (). The magnitude and
mass and body weight during leptin treatment. Those time-course of the weight change in this study is not
studies either showed no change in bone density (, reported, so it is not possible to determine any

doi: 10.1210/er.2017-00226 https://academic.oup.com/edrv 953


REVIEW

temporal relationship between weight loss and PTH of specific pathologies. Therefore, circumspection is
levels. The Foo study also showed a reduction in the needed in defining leptin’s role in any particular
ratio of RANKL to osteoprotegerin, suggesting an context.
antiresorptive effect of leptin, possibly secondary to There are limitations in the experimental models
inhibition of PTH release. However the inconsistency used to demonstrate the direct and indirect effects of
of the changes in PTH levels and bone turnover leptin on bone. Leptin in the circulation crosses the
markers in the other human studies means that blood-brain barrier, so it can affect hypothalamic
changes in PTH cannot be regarded as an established and pituitary function. The latter is likely to be
mechanism of the leptin effects on bone. particularly relevant to the effects of systemic leptin
Two studies in women with hypothalamic administration on women with hypothalamic

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


amenorrhea showed profound changes in sex hor- amenorrhea, in whom changes in gonadal, thyroid,
mone levels, thyroid hormones and the IGF system. and adrenal function and in circulating levels of
In the study of Chou et al. (), % of the growth hormone and IGF were observed after
amenorrheic women treated with leptin had a return systemic treatment (Table ). However, the reverse
of menstruation. In the Welt et al. () study, es- is also the case, because there is reabsorption of the
tradiol levels increased threefold after  months of cerebrospinal fluid into the blood (). Maness
leptin treatment, thyroid hormone levels by % to et al. () showed that the amount of leptin in the
%, and IGF by %. All these changes would be peripheral circulation after injection into the ce-
expected to have beneficial effects on bone formation rebral ventricles is comparable to levels seen
and bone mass.  minutes after intravenous administration. Thus,
Despite some inconsistencies between these central leptin infusion at  ng/h (i.e., similar to or
studies, their results are entirely consistent with the less than dosages used in central leptin adminis-
body of animal data and indicate that the systemic tration studies) produces detectable levels of leptin
administration of leptin to either obese or un- in the circulation and results in leptin receptor
derweight subjects results in weight loss, with either activation in peripheral cells such as thymocytes
preservation of bone density or increased bone (). Therefore, some of the short-term effects of
mass. In almost all other contexts, weight loss is centrally administered leptin might be mediated by
associated with reduction in bone mass. Therefore, direct peripheral effects. Furthermore, rats with
these studies reinforce the body of evidence in- sustained central leptin expression produced by a
dicating that systemic administration of leptin is single ICV injection of a recombinant adenoasso-
anabolic to bone. These studies do not resolve ciated virus vector encoding leptin have marked
whether this is a direct or indirect effect. The peripheral hypoleptinemia and hypoinsulinemia
changes in sex hormones, thyroid hormones, and (). Weight loss alone can reduce circulating
the IGF system demonstrated by Welt et al. () leptin by % (), but pair-feeding studies indicate
and Chou et al. () indicate a high likelihood that, that central administration of leptin reduces its
in humans, some of leptin’s effects on bone are circulating levels independent of the weight loss
indirectly mediated through these hormones. (). The hypoinsulinemia that follows central
leptin administration is likely to be accompanied by
reductions in amylin and preptin, which are also
Integrating the Direct and Indirect Effects of secreted by the pancreatic b cell. All these peptides
Leptin on the Skeleton have anabolic effects on bone (–). Thus, the
direct anabolic effects on bone of this cluster of
The literature reviewed above indicates that leptin has peptides and of leptin itself are substantially reduced
direct anabolic effects on bone cells or can influence by central leptin administration. A diagram sum-
bone indirectly in a number of ways: via its actions in marizing these possible pathways for leptin effects on
the CNS, which are catabolic to vertebral trabecular bone is shown in Fig. .
bone; via changes in weight (catabolic); and via effects When assessing studies of centrally and periph-
on the production of other hormones, which are erally administered leptin, we need to consider how
usually anabolic. Leptin’s role in bone physiology will physiological the dosages used are. Philbrick et al. ()
be determined by the interaction and balance of these demonstrated that a systemic infusion rate of  ng/h
conflicting effects. It is possible that this balance (equivalent to . mg/d) achieves physiological cir-
might vary between species; the SNS-mediated effects culating leptin levels. By this standard, many of the
are well established in mice but not in humans, studies of systemic administration set out in Table 
where adrenergic blockers have little effect on the used substantially supraphysiological leptin dosages.
skeleton. The balance of leptin’s anabolic and cat- However, Maness et al. () showed that the brain/
abolic effects might also vary according to factors serum ratios of leptin produced by ICV injection are
such as body weight, baseline circulating leptin  times higher than those produced by intravenous
levels, the level of leptin resistance, and the presence administration, so even delivery of leptin into the

954 Reid et al Effects of Leptin on the Skeleton Endocrine Reviews, December 2018, 39(6):938–959
REVIEW

Figure 7. Pathways by
which leptin, originating in
Adipose tissue adipose tissue, bone
Hypothalamus marrow adipocytes, and
osteoblasts, influences
bone. The width of the
yellow arrows indicates the
Marrow Sympathetic leptin concentrations
adipocytes nervous found in bone and in the
Leptin in system
CNS. [© 2018 Illustration
circulation
Presentation ENDOCRINE

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


Weight loss
SOCIETY].
– Blood leptin
Blood insulin
Euglycemia
– Pituitary
Leptin
in bone
LepRb
Eugonadism
+ Euthyroidism
Eucortisolism
Osteoblast Chondrocyte IGF1

Proliferation, differentiation, Proliferation, differentiation,


protein synthesis, mineralization, growth plate thickness
OPG; RANKL © 2018 Illustration Presentation ENDOCRINE SOCIETY

cerebral ventricles at  ng/h might produce brain levels to reproduce these findings. Thus, the balance of
much higher than would occur physiologically. the central and peripheral effects of leptin on bone
Several lines of evidence suggest that the direct remains an area of substantial controversy, reso-
effects of leptin on bone are likely to be dominant in lution of which will require additional experi-
most circumstances. Leptin is produced outside the mental work.
CNS and is present in the circulation in concen- Whether the balance of central and peripheral
trations  to  times higher than those in the effects of leptin is similar in different models,
cerebrospinal fluid. It is produced in bone marrow particularly in humans, is unknown. In fact, the
adipocytes, chondrocytes (), and cells of the largest deficit in our knowledge of the effects of
osteoblast lineage (), so its concentrations leptin on the skeleton is in human studies, and
around bone cells might be even higher than in the there is a need for substantial work in this regard. A
circulation. The elegant dose-response leptin in- careful study of the dose response of leptin
fusion studies that Philbrick et al. () carried out in treatment in patients who are leptin-deficient al-
ob/ob mice provide persuasive evidence that leptin ready receiving sex hormone replacement could
directly stimulates bone cells, increasing bone address leptin’s effects on the skeleton in humans,
formation and producing robust changes in Jak/ but the rarity of such patients is a real challenge for
Stat signaling genes in bone and bone cell–related such studies. The fact that leptin and body weight
genes at leptin infusion rates that do not activate are positively related to bone density and inversely
Jak/Stat signaling in the hypothalamus or produce related to fracture risk suggests that the direct
its centrally mediated effects on energy meta- peripheral effects predominate in humans, and this
bolism. This evidence is supported by the finding supposition is supported by the absence of evi-
that transferring db/db bone marrow into wild-type dence that b-blockers significantly affect human
mice reproduces the skeletal phenotype of the db/ bone health. The absence of a clear bone pheno-
db mouse without evidence of any change in the type in the small number of human cases of severe
CNS actions of leptin (), indicating that the loss long-term leptin deficiency that have been de-
of leptin signaling in bone alone is adequate to scribed suggests that neither peripheral nor cen-
reproduce the bone effects of leptin deficiency. tral effects are critical to human bone health.
However, Shi et al. () found contrary results, that However, the demonstration of a neural pathway
selective knockout of osteoblastic leptin receptors involving the SNS that permits the CNS to in-
did not affect the bone phenotype in mice. Also, the fluence bone formation is a fascinating develop-
Philbrick et al. () work has only just appeared, ment in this area of research. It remains possible
and other groups have not yet had the opportunity that in some contexts this pathway might be

doi: 10.1210/er.2017-00226 https://academic.oup.com/edrv 955


REVIEW

physiologically or pathologically important in appreciation of the regulatory interactions oc-


human health. Recognition of the interactions curring in the skeleton, particularly the interplay
between leptin and bone has broadened our between energy and bone homeostasis.

References
1. Reid IR. Fat and bone. Arch Biochem Biophys. 2010; 12. Bassilana F, Susa M, Keller HJ, Halleux C. Human 28. Myers MG Jr, Heymsfield SB, Haft C, Kahn BB,
503(1):20–27. mesenchymal stem cells undergoing osteo- Laughlin M, Leibel RL, Tschöp MH, Yanovski JA.

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


2. Johansson H, Kanis JA, Odén A, McCloskey E, genic differentiation express leptin and func- Challenges and opportunities of defining clinical
Chapurlat RD, Christiansen C, Cummings SR, Diez- tional leptin receptor. J Bone Miner Res. 2000; leptin resistance. Cell Metab. 2012;15(2):150–156.
Perez A, Eisman JA, Fujiwara S, Glüer CC, Goltzman 15(suppl 1):S378. 29. Jung CH, Kim M-S. Molecular mechanisms of
D, Hans D, Khaw KT, Krieg MA, Kröger H, LaCroix 13. Reseland JE, Syversen U, Bakke I, Qvigstad G, Eide central leptin resistance in obesity. Arch Pharm Res.
AZ, Lau E, Leslie WD, Mellström D, Melton LJ III, LG, Hjertner O, Gordeladze JO, Drevon CA. Leptin 2013;36(2):201–207.
O’Neill TW, Pasco JA, Prior JC, Reid DM, Rivadeneira is expressed in and secreted from primary cultures 30. Rizwan MZ, Mehlitz S, Grattan DR, Tups A. Tem-
F, van Staa T, Yoshimura N, Zillikens MC. A meta- of human osteoblasts and promotes bone min- poral and regional onset of leptin resistance in diet-
analysis of the association of fracture risk and body eralization. J Bone Miner Res. 2001;16(8): induced obese mice. J Neuroendocrinol. 2017;29(10):
mass index in women. J Bone Miner Res. 2014;29(1): 1426–1433. e12481.
223–233. 14. Kume K, Satomura K, Nishisho S, Kitaoka E, 31. El-Haschimi K, Pierroz DD, Hileman SM, Bjørbaek C,
3. Reid IR. Relationships between fat and bone. Yamanouchi K, Tobiume S, Nagayama M. Potential Flier JS. Two defects contribute to hypothalamic
Osteoporos Int. 2008;19(5):595–606. role of leptin in endochondral ossification. J Histochem leptin resistance in mice with diet-induced obesity.
4. De Laet C, Kanis JA, Odén A, Johanson H, Johnell O, Cytochem. 2002;50(2):159–169. J Clin Invest. 2000;105(12):1827–1832.
Delmas P, Eisman JA, Kroger H, Fujiwara S, Garnero 15. Park H-K, Ahima RS. Physiology of leptin: energy 32. Banks WA, Kastin AJ, Huang W, Jaspan JB, Maness
P, McCloskey EV, Mellstrom D, Melton LJ III, homeostasis, neuroendocrine function and meta- LM. Leptin enters the brain by a saturable sys-
Meunier PJ, Pols HAP, Reeve J, Silman A, bolism. Metabolism. 2015;64(1):24–34. tem independent of insulin. Peptides. 1996;17(2):
Tenenhouse A. Body mass index as a predictor of 16. Licinio J, Negrão AB, Mantzoros C, Kaklamani V, 305–311.
fracture risk: a meta-analysis. Osteoporos Int. 2005; Wong ML, Bongiorno PB, Negro PP, Mulla A, 33. Caro JF, Kolaczynski JW, Nyce MR, Ohannesian JP,
16(11):1330–1338. Veldhuis JD, Cearnal L, Flier JS, Gold PW. Sex dif- Opentanova I, Goldman WH, Lynn RB, Zhang PL,
5. Ensrud KE, Lipschutz RC, Cauley JA, Seeley D, Nevitt ferences in circulating human leptin pulse ampli- Sinha MK, Considine RV. Decreased cerebrospinal-
MC, Scott J, Orwoll ES, Genant HK, Cummings SR; tude: clinical implications. J Clin Endocrinol Metab. fluid/serum leptin ratio in obesity: a possible
Study of Osteoporotic Fractures Research Group. 1998;83(11):4140–4147. mechanism for leptin resistance. Lancet. 1996;
Body size and hip fracture risk in older women: 17. Chan JL, Heist K, DePaoli AM, Veldhuis JD, 348(9021):159–161.
a prospective study. Am J Med. 1997;103(4):274–280. Mantzoros CS. The role of falling leptin levels in the 34. Nam SY, Kratzsch J, Kim KW, Kim KR, Lim SK,
6. Schott AM, Cormier C, Hans D, Favier F, Hausherr E, neuroendocrine and metabolic adaptation to short- Marcus C. Cerebrospinal fluid and plasma con-
Dargent-Molina P, Delmas PD, Ribot C, Sebert JL, term starvation in healthy men. J Clin Invest. 2003; centrations of leptin, NPY, and alpha-MSH in obese
Breart G, Meunier PJ. How hip and whole-body 111(9):1409–1421. women and their relationship to negative energy
bone mineral density predict hip fracture in elderly 18. Rosenbaum M, Leibel RL. Clinical review 107: role of balance. J Clin Endocrinol Metab. 2001;86(10):
women: the EPIDOS Prospective Study. Osteoporos gonadal steroids in the sexual dimorphisms in body 4849–4853.
Int. 1998;8(3):247–254. composition and circulating concentrations of 35. Philbrick KA, Wong CP, Branscum AJ, Turner RT,
7. Lau EMC, Chan YH, Chan M, Woo J, Griffith J, Chan leptin. J Clin Endocrinol Metab. 1999;84:1784–1789. Iwaniec UT. Leptin stimulates bone formation in
HHL, Leung PC. Vertebral deformity in Chinese 19. Ahima RS, Osei SY. Leptin signaling. Physiol Behav. ob/ob mice at doses having minimal impact on
men: prevalence, risk factors, bone mineral density, 2004;81(2):223–241. energy metabolism. J Endocrinol. 2017;232(3):
and body composition measurements. Calcif Tissue 20. Flak JN, Myers MG Jr. Minireview: CNS mechanisms 461–474.
Int. 2000;66(1):47–52. of leptin action. Mol Endocrinol. 2016;30(1):3–12. 36. Ducy P, Amling M, Takeda S, Priemel M, Schilling AF,
8. Lacombe J, Cairns BJ, Green J, Reeves GK, Beral V, 21. Tartaglia LA. The leptin receptor. J Biol Chem. 1997; Beil FT, Shen J, Vinson C, Rueger JM, Karsenty G.
Armstrong MEG; Million Women Study collabo- 272(10):6093–6096. Leptin inhibits bone formation through a hypo-
rators. The effects of age, adiposity, and physical 22. de Luca C, Kowalski TJ, Zhang Y, Elmquist JK, Lee C, thalamic relay: a central control of bone mass. Cell.
activity on the risk of seven site-specific fractures in Kilimann MW, Ludwig T, Liu S-M, Chua SC Jr. 2000;100(2):197–207.
postmenopausal women. J Bone Miner Res. 2016; Complete rescue of obesity, diabetes, and infertility 37. Steppan CM, Crawford DT, Chidsey-Frink KL, Ke H,
31(8):1559–1568. in db/db mice by neuron-specific LEPR-B transgenes. Swick AG. Leptin is a potent stimulator of bone
9. Compston JE, Flahive J, Hosmer DW, Watts NB, Siris J Clin Invest. 2005;115(12):3484–3493. growth in ob/ob mice. Regul Pept. 2000;92(1–3):73–78.
ES, Silverman S, Saag KG, Roux C, Rossini M, 23. Bjørbaek C, Elmquist JK, Michl P, Ahima RS, van 38. Hamrick MW, Pennington C, Newton D, Xie D,
Pfeilschifter J, Nieves JW, Netelenbos JC, March L, Bueren A, McCall AL, Flier JS. Expression of leptin Isales C. Leptin deficiency produces contrasting
LaCroix AZ, Hooven FH, Greenspan SL, Gehlbach receptor isoforms in rat brain microvessels. Endo- phenotypes in bones of the limb and spine. Bone.
SH, Dı́ez-Pérez A, Cooper C, Chapurlat RD, Boonen crinology. 1998;139(8):3485–3491. 2004;34(3):376–383.
S, Anderson FA Jr, Adami S, Adachi JD; GLOW 24. Dalamaga M, Chou SH, Shields K, Papageorgiou P, 39. Hamrick MW, Della-Fera MA, Choi YH, Pennington
Investigators. Relationship of weight, height, and Polyzos SA, Mantzoros CS. Leptin at the intersection C, Hartzell D, Baile CA. Leptin treatment induces
body mass index with fracture risk at different sites of neuroendocrinology and metabolism: current loss of bone marrow adipocytes and increases bone
in postmenopausal women: the Global Longitudi- evidence and therapeutic perspectives. Cell Metab. formation in leptin-deficient ob/ob mice. J Bone
nal Study of Osteoporosis in Women (GLOW). 2013;18(1):29–42. Miner Res. 2005;20(6):994–1001.
J Bone Miner Res. 2014;29(2):487–493. 25. Haynes WG, Morgan DA, Walsh SA, Mark AL, Sivitz 40. Kishida Y, Hirao M, Tamai N, Nampei A, Fujimoto T,
10. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, WI. Receptor-mediated regional sympathetic nerve Nakase T, Shimizu N, Yoshikawa H, Myoui A. Leptin
Friedman JM. Positional cloning of the mouse obese activation by leptin. J Clin Invest. 1997;100(2):270–278. regulates chondrocyte differentiation and matrix
gene and its human homologue. Nature. 1994; 26. Scarpace PJ, Matheny M, Pollock BH, Tümer N. maturation during endochondral ossification. Bone.
372(6505):425–432. Leptin increases uncoupling protein expression and 2005;37(5):607–621.
11. Laharrague P, Larrouy D, Fontanilles AM, Truel N, energy expenditure. Am J Physiol. 1997;273(1 Pt 1): 41. Baldock PA, Sainsbury A, Allison S, Lin EJD, Couzens
Campfield A, Tenenbaum R, Galitzky J, Corberand E226–E230. M, Boey D, Enriquez R, During M, Herzog H,
JX, Pénicaud L, Casteilla L. High expression of leptin 27. Mantzoros CS, Flier JS. Editorial: leptin as a thera- Gardiner EM. Hypothalamic control of bone for-
by human bone marrow adipocytes in primary peutic agent--trials and tribulations. J Clin Endocrinol mation: distinct actions of leptin and y2 receptor
culture. FASEB J. 1998;12(9):747–752. Metab. 2000;85(11):4000–4002. pathways. J Bone Miner Res. 2005;20(10):1851–1857.

956 Reid et al Effects of Leptin on the Skeleton Endocrine Reviews, December 2018, 39(6):938–959
REVIEW

42. Ealey KN, Fonseca D, Archer MC, Ward WE. Bone concentrations and bone mineral density, and hyperparathyroidism. Singapore Med J. 2011;
abnormalities in adolescent leptin-deficient mice. biochemical markers of bone turnover in adult 52(12):890–893.
Regul Pept. 2006;136(1–3):9–13. men. J Clin Endocrinol Metab. 2001;86(11): 75. Grey AB, Evans MC, Stapleton JP, Reid IR. Body
43. Iwaniec UT, Boghossian S, Lapke PD, Turner RT, 5273–5276. weight and bone mineral density in post-
Kalra SP. Central leptin gene therapy corrects 61. Ho-Pham LT, Lai TQ, Nguyen UDT, Bui QV, Nguyen menopausal women with primary hyperpara-
skeletal abnormalities in leptin-deficient ob/ob TV. Delineating the relationship between leptin, fat thyroidism. Ann Intern Med. 1994;121(10):
mice. Peptides. 2007;28(5):1012–1019. mass, and bone mineral density: a mediation 745–749.
44. Turner RT, Kalra SP, Wong CP, Philbrick KA, analysis. Calcif Tissue Int. 2017;100(1):13–19. 76. Hoang D, Broer N, Roman SA, Yao X, Abitbol N, Li F,
Lindenmaier LB, Boghossian S, Iwaniec UT. Pe- 62. Javaid MK, Godfrey KM, Taylor P, Robinson SM, Sosa JA, Sue GR, DeWan AT, Wong ML, Licinio J,
ripheral leptin regulates bone formation. J Bone Crozier SR, Dennison EM, Robinson JS, Breier BR, Simpson C, Li AY, Pizzoferrato N, Narayan D. Leptin
Miner Res. 2013;28(1):22–34. Arden NK, Cooper C. Umbilical cord leptin predicts signaling and hyperparathyroidism: clinical and
45. Lorentzon R, Alehagen U, Boquist L. Osteopenia in neonatal bone mass. Calcif Tissue Int. 2005;76(5): genetic associations. J Am Coll Surg. 2014;218(6):

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


mice with genetic diabetes. Diabetes Res Clin Pract. 341–347. 1239–1250.e4.
1986;2(3):157–163. 63. Rhie YJ, Lee KH, Chung SC, Kim HS, Kim DH. Effects 77. Matsunuma A, Kawane T, Maeda T, Hamada S,
46. Takeshita N, Mutoh S, Yamaguchi I; Basic Research of body composition, leptin, and adiponectin on Horiuchi N. Leptin corrects increased gene
Group. Osteopenia in genetically diabetic DB/DB bone mineral density in prepubertal girls. J Korean expression of renal 25-hydroxyvitamin D3-1 alpha-
mice and effects of 1a-hydroxyvitamin D3 on the Med Sci. 2010;25(8):1187–1190. hydroxylase and -24-hydroxylase in leptin-
osteopenia. Life Sci. 1995;56(13):1095–1101. 64. Huang KC, Cheng WC, Yen RF, Tsai KS, Tai TY, Yang deficient, ob/ob mice. Endocrinology. 2004;
47. Williams GA, Callon KE, Watson M, Costa JL, Ding Y, WS. Lack of independent relationship between 145(3):1367–1375.
Dickinson M, Wang Y, Naot D, Reid IR, Cornish J. plasma adiponectin, leptin levels and bone density 78. Tsuji K, Maeda T, Kawane T, Matsunuma A,
Skeletal phenotype of the leptin receptor-deficient in nondiabetic female adolescents. Clin Endocrinol Horiuchi N. Leptin stimulates fibroblast growth
db/db mouse. J Bone Miner Res. 2011;26(8): (Oxf). 2004;61(2):204–208. factor 23 expression in bone and suppresses renal
1698–1709. 65. Yamauchi M, Sugimoto T, Yamaguchi T, Nakaoka 1alpha,25-dihydroxyvitamin D3 synthesis in leptin-
48. Foldes J, Shih MS, Levy J. Bone structure and calcium D, Kanzawa M, Yano S, Ozuru R, Sugishita T, deficient mice. J Bone Miner Res. 2010;25(8):
metabolism in obese Zucker rats. Int J Obes Relat Chihara K. Plasma leptin concentrations are 1711–1723.
Metab Disord. 1992;16(2):95–102. associated with bone mineral density and 79. Hoang D, Broer N, Sosa JA, Abitbol N, Yao X, Li F,
49. Mathey J, Horcajada-Molteni MN, Chanteranne B, the presence of vertebral fractures in post- Rivera-Molina F, Toomre DK, Roman SA, Sue G, Kim
Picherit C, Puel C, Lebecque P, Cubizoles C, Davicco menopausal women. Clin Endocrinol (Oxf). 2001; S, Li AY, Callender GG, Simpson C, Narayan D.
MJ, Coxam V, Barlet JP. Bone mass in obese diabetic 55(3):341–347. Leptin is produced by parathyroid glands and
Zucker rats: influence of treadmill running. Calcif 66. Schett G, Kiechl S, Bonora E, Redlich K, Woloszczuk stimulates parathyroid hormone secretion. Ann
Tissue Int. 2002;70(4):305–311. W, Oberhollenzer F, Jocher J, Dorizzi R, Muggeo M, Surg. 2017;266(6):1075–1083.
50. Tamasi JA, Arey BJ, Bertolini DR, Feyen JHM. Smolen J, Willeit J. Serum leptin level and the risk of 80. Mirza MA, Alsiö J, Hammarstedt A, Erben RG,
Characterization of bone structure in leptin nontraumatic fracture. Am J Med. 2004;117(12): Michaëlsson K, Tivesten A, Marsell R, Orwoll E,
receptor–deficient Zucker (fa/fa) rats. J Bone Miner 952–956. Karlsson MK, Ljunggren O, Mellström D, Lind L,
Res. 2003;18(9):1605–1611. 67. Barbour KE, Zmuda JM, Boudreau R, Strotmeyer ES, Ohlsson C, Larsson TE. Circulating fibroblast growth
51. Simha V, Zerwekh JE, Sakhaee K, Garg A. Effect of Horwitz MJ, Evans RW, Kanaya AM, Harris TB, Bauer factor-23 is associated with fat mass and dyslipi-
subcutaneous leptin replacement therapy on bone DC, Cauley JA. Adipokines and the risk of fracture demia in two independent cohorts of elderly in-
metabolism in patients with generalized lipodys- in older adults. J Bone Miner Res. 2011;26(7): dividuals. Arterioscler Thromb Vasc Biol. 2011;31(1):
trophy. J Clin Endocrinol Metab. 2002;87(11): 1568–1576. 219–227.
4942–4945. 68. Shaarawy M, Abassi AF, Hassan H, Salem ME. Re- 81. Billington EO, Gamble GD, Bristow S, Reid IR. Serum
52. Ozata M. Different presentation of bone mass in lationship between serum leptin concentrations phosphate is related to adiposity in healthy adults:
mice and humans with congenital leptin deficiency. and bone mineral density as well as biochemical cross-sectional assessment of three independent
J Clin Endocrinol Metab. 2002;87(2):951. markers of bone turnover in women with post- cohorts. Eur J Clin Invest. 2017;47(7):486–493.
53. Himms-Hagen J. Physiological roles of the leptin menopausal osteoporosis. Fertil Steril. 2003;79(4): 82. Saini RK, Kaneko I, Jurutka PW, Forster R, Hsieh A,
endocrine system: differences between mice and 919–924. Hsieh J-C, Haussler MR, Whitfield GK. 1,25-
humans. Crit Rev Clin Lab Sci. 1999;36(6):575–655. 69. Blain H, Vuillemin A, Guillemin F, Durant R, Hanesse dihydroxyvitamin D(3) regulation of fibroblast
54. Farooqi IS, Jebb SA, Langmack G, Lawrence E, B, de Talance N, Doucet B, Jeandel C. Serum leptin growth factor-23 expression in bone cells: evidence
Cheetham CH, Prentice AM, Hughes IA, McCamish level is a predictor of bone mineral density in for primary and secondary mechanisms modulated
MA, O’Rahilly S. Effects of recombinant leptin postmenopausal women. J Clin Endocrinol Metab. by leptin and interleukin-6. Calcif Tissue Int. 2013;
therapy in a child with congenital leptin deficiency. 2002;87(3):1030–1035. 92(4):339–353.
N Engl J Med. 1999;341(12):879–884. 70. Scariano JK, Garry PJ, Montoya GD, Chandani AK, 83. Matsunuma A, Horiuchi N. Leptin attenuates gene
55. Weiss LA, Barrett-Connor E, von Mühlen D, Clark P. Wilson JM, Baumgartner RN. Serum leptin levels, expression for renal 25-hydroxyvitamin D3-1alpha-
Leptin predicts BMD and bone resorption in older bone mineral density and osteoblast alkaline hydroxylase in mice via the long form of the leptin
women but not older men: the Rancho Bernardo phosphatase activity in elderly men and women. receptor. Arch Biochem Biophys. 2007;463(1):
study. J Bone Miner Res. 2006;21(5):758–764. Mech Ageing Dev. 2003;124(3):281–286. 118–127.
56. Reid IR, Richards JB. Adipokine effects on bone. Clin 71. Prouteau S, Benhamou L, Courteix D. Relationships 84. Bartell SM, Rayalam S, Ambati S, Gaddam DR,
Rev Bone Miner Metab. 2009;7(3):240–248. between serum leptin and bone markers during Hartzell DL, Hamrick M, She JX, Della-Fera MA, Baile
57. Goulding A, Taylor RW. Plasma leptin values in stable weight, weight reduction and weight regain CA. Central (ICV) leptin injection increases bone
relation to bone mass and density and to dynamic in male and female judoists. Eur J Endocrinol. 2006; formation, bone mineral density, muscle mass, se-
biochemical markers of bone resorption and for- 154(3):389–395. rum IGF-1, and the expression of osteogenic genes
mation in postmenopausal women. Calcif Tissue Int. 72. Grethen E, Hill KM, Jones R, Cacucci BM, Gupta CE, in leptin-deficient ob/ob mice. J Bone Miner Res.
1998;63(6):456–458. Acton A, Considine RV, Peacock M. Serum leptin, 2011;26(8):1710–1720.
58. Mpalaris V, Anagnostis P, Anastasilakis AD, Goulis parathyroid hormone, 1,25-dihydroxyvitamin D, 85. Pogoda P, Egermann M, Schnell JC, Priemel M,
DG, Doumas A, Iakovou I. Serum leptin, adiponectin fibroblast growth factor 23, bone alkaline phos- Schilling AF, Alini M, Schinke T, Rueger JM,
and ghrelin concentrations in post-menopausal phatase, and sclerostin relationships in obesity. J Clin Schneider E, Clarke I, Amling M. Leptin in-
women: is there an association with bone mineral Endocrinol Metab. 2012;97(5):1655–1662. hibits bone formation not only in rodents, but
density? Maturitas. 2016;88:32–36. 73. Billington EO, Gamble GD, Reid IR. Parathyroid also in sheep. J Bone Miner Res. 2006;21(10):
59. Ruhl CE, Everhart JE. Relationship of serum leptin hormone reflects adiposity and cardiometabolic 1591–1599.
concentration with bone mineral density in the indices but not bone density in normal men. 86. Takeda S, Elefteriou F, Levasseur R, Liu X, Zhao L,
United States population. J Bone Miner Res. 2002; Bonekey Rep. 2016;5:852. Parker KL, Armstrong D, Ducy P, Karsenty G. Leptin
17(10):1896–1903. 74. Bhadada SK, Bhansali A, Shah VN, Rao DS. Changes regulates bone formation via the sympathetic
60. Sato M, Takeda N, Sarui H, Takami R, Takami K, in serum leptin and adiponectin concentra- nervous system. Cell. 2002;111(3):305–317.
Hayashi M, Sasaki A, Kawachi S, Yoshino K, tions and insulin resistance after curative para- 87. Ruffin M, Nicolaidis S. Electrical stimulation of the
Yasuda K. Association between serum leptin thyroidectomy in moderate to severe primary ventromedial hypothalamus enhances both fat

doi: 10.1210/er.2017-00226 https://academic.oup.com/edrv 957


REVIEW

utilization and metabolic rate that precede and obese (ob/ob) mouse. Endocrinology. 1993;132(5): 116. Lee YJ, Park JH, Ju SK, You KH, Ko JS, Kim HM. Leptin
parallel the inhibition of feeding behavior. Brain Res. 1939–1944. receptor isoform expression in rat osteoblasts and
1999;846(1):23–29. 103. Baldock PA, Lee NJ, Driessler F, Lin S, Allison S, their functional analysis. FEBS Lett. 2002;528(1–3):
88. Elefteriou F, Ahn JD, Takeda S, Starbuck M, Yang X, Stehrer B, Lin EJ, Zhang L, Enriquez RF, Wong IP, 43–47.
Liu X, Kondo H, Richards WG, Bannon TW, Noda McDonald MM, During M, Pierroz DD, Slack K, Shi 117. Burguera B, Brunetto A, Garcia-Ocana A, Teijeiro R,
M, Clement K, Vaisse C, Karsenty G. Leptin regu- YC, Yulyaningsih E, Aljanova A, Little DG, Ferrari SL, Esplen J, Thomas T, Couce ME, Zhao A. Leptin
lation of bone resorption by the sympathetic Sainsbury A, Eisman JA, Herzog H. Neuropeptide Y increases proliferation of human steosarcoma cells
nervous system and CART. Nature. 2005;434(7032): knockout mice reveal a central role of NPY in the through activation of PI(3)-K and MAPK pathways.
514–520. coordination of bone mass to body weight. PLoS Med Sci Monit. 2006;12(11):BR341–BR349.
89. Pierroz DD, Bonnet N, Bianchi EN, Bouxsein ML, One. 2009;4(12):e8415. 118. Iwaniec UT, Shearon CC, Heaney RP, Cullen DM,
Baldock PA, Rizzoli R, Ferrari SL. Deletion of 104. Wong IP, Nguyen AD, Khor EC, Enriquez RF, Eisman Yee JA. Leptin increases number of bone nodules
b-adrenergic receptor 1, 2, or both leads to dif- JA, Sainsbury A, Herzog H, Baldock PA. Neuro- in vitro. Bone. 1998;13(5, suppl):S212.

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


ferent bone phenotypes and response to me- peptide Y is a critical modulator of leptin’s regu- 119. Thomas T, Gori F, Khosla S, Jensen MD, Burguera B,
chanical stimulation. J Bone Miner Res. 2012;27(6): lation of cortical bone. J Bone Miner Res. 2013;28: Riggs BL. Leptin acts on human marrow stromal
1252–1262. 886–898. cells to enhance differentiation to osteoblasts and
90. Shi Y, Yadav VK, Suda N, Liu XS, Guo XE, Myers MG 105. Hamrick MW, Ding KH, Ponnala S, Ferrari SL, to inhibit differentiation to adipocytes. Endocri-
Jr, Karsenty G. Dissociation of the neuronal regu- Isales CM. Caloric restriction decreases cortical nology. 1999;140(4):1630–1638.
lation of bone mass and energy metabolism by bone mass but spares trabecular bone in the 120. Zhang J, Li T, Xu L, Li W, Cheng M, Zhuang J, Chen Y,
leptin in vivo. Proc Natl Acad Sci USA. 2008;105(51): mouse skeleton: implications for the regulation of Xu W. Leptin promotes ossification through mul-
20529–20533. bone mass by body weight. J Bone Miner Res. 2008; tiple ways of bone metabolism in osteoblast: a pilot
91. Gallardo N, Bonzón-Kulichenko E, Fernández- 23:870–878. study. Gynecol Endocrinol. 2013;29(8):758–762.
Agulló T, Moltó E, Gómez-Alonso S, Blanco P, 106. Walther DJ, Peter JU, Bashammakh S, Hörtnagl H, 121. Grundberg E, Brändström H, Lam KCL, Gurd S, Ge B,
Carrascosa JM, Ros M, Andrés A. Tissue-specific Voits M, Fink H, Bader M. Synthesis of serotonin Harmsen E, Kindmark A, Ljunggren O, Mallmin H,
effects of central leptin on the expression of by a second tryptophan hydroxylase isoform. Sci- Nilsson O, Pastinen T. Systematic assessment of the
genes involved in lipid metabolism in liver and ence. 2003;299(5603):76. human osteoblast transcriptome in resting and
white adipose tissue. Endocrinology. 2007;148(12): 107. Yadav VK, Oury F, Suda N, Liu ZW, Gao XB, induced primary cells. Physiol Genomics. 2008;33(3):
5604–5610. Confavreux C, Klemenhagen KC, Tanaka KF, 301–311.
92. Hamrick MW, Della Fera MA, Choi YH, Hartzell D, Gingrich JA, Guo XE, Tecott LH, Mann JJ, Hen R, 122. Khan SN, DuRaine G, Virk SS, Fung J, Rowland DJ,
Pennington C, Baile CA. Injections of leptin into rat Horvath TL, Karsenty G. A serotonin-dependent Reddi AH, Lee MA. The temporal role of leptin
ventromedial hypothalamus increase adipocyte mechanism explains the leptin regulation of bone within fracture healing and the effect of local ap-
apoptosis in peripheral fat and in bone marrow. Cell mass, appetite, and energy expenditure. Cell. 2009; plication of recombinant leptin on fracture healing.
Tissue Res. 2007;327(1):133–141. 138(5):976–989. J Orthop Trauma. 2013;27(11):656–662.
93. Veldhuis-Vlug AG, Rosen CJ. Clinical implications of 108. Yadav VK, Oury F, Tanaka KF, Thomas T, Wang Y, 123. Beil FT, Barvencik F, Gebauer M, Beil B, Pogoda P,
bone marrow adiposity. J Intern Med. 2018;283(2): Cremers S, Hen R, Krust A, Chambon P, Karsenty G. Rueger JM, Ignatius A, Schinke T, Amling M. Effects
121–139. Leptin-dependent serotonin control of appetite: of increased bone formation on fracture healing in
94. Schlienger RG, Kraenzlin ME, Jick SS, Meier CR. Use temporal specificity, transcriptional regulation, and mice. J Trauma. 2011;70(4):857–862.
of beta-blockers and risk of fractures. JAMA. 2004; therapeutic implications [published correction 124. Holloway WR, Collier FM, Aitken CJ, Myers DE,
292(11):1326–1332. appears in J Exp Med. 2011;208(2):413]. J Exp Med. Hodge JM, Malakellis M, Gough TJ, Collier GR,
95. Rejnmark L, Vestergaard P, Mosekilde L. Treatment 2011;208(1):41–52. Nicholson GC. Leptin inhibits osteoclast generation.
with beta-blockers, ACE inhibitors, and calcium- 109. Cornish J, Callon KE, Bava U, Lin C, Naot D, Hill BL, J Bone Miner Res. 2002;17(2):200–209.
channel blockers is associated with a reduced Grey AB, Broom N, Myers DE, Nicholson GC, Reid IR. 125. Burguera B, Hofbauer LC, Thomas T, Gori F, Evans
fracture risk: a nationwide case-control study. Leptin directly regulates bone cell function in vitro GL, Khosla S, Riggs BL, Turner RT. Leptin reduces
J Hypertens. 2006;24(3):581–589. and reduces bone fragility in vivo. J Endocrinol. 2002; ovariectomy-induced bone loss in rats. Endocri-
96. de Vries F, Souverein PC, Cooper C, Leufkens HGM, 175(2):405–415. nology. 2001;142(8):3546–3553.
van Staa TP. Use of beta-blockers and the risk of 110. Lee YJ, Park JH, Ko JS, Kim HM. Expression of signal- 126. Lamghari M, Tavares L, Camboa N, Barbosa MA.
hip/femur fracture in the United Kingdom and The transducing leptin receptors on rat osteoblasts Leptin effect on RANKL and OPG expression in
Netherlands. Calcif Tissue Int. 2007;80(2):69–75. indicates a direct involvement of leptin in the MC3T3-E1 osteoblasts. J Cell Biochem. 2006;98(5):
97. Yang S, Nguyen ND, Center JR, Eisman JA, Nguyen regulation of bone formation. J Bone Miner Res. 1123–1129.
TV. Association between beta-blocker use and 2001;16(suppl 1):S495. 127. Maor G, Rochwerger M, Segev Y, Phillip M. Leptin
fracture risk: the Dubbo Osteoporosis Epidemiology 111. Enjuanes A, Supervı́a A, Nogués X, Dı́ez-Pérez A. acts as a growth factor on the chondrocytes of
Study. Bone. 2011;48(3):451–455. Leptin receptor (OB-R) gene expression in human skeletal growth centers. J Bone Miner Res. 2002;17(6):
98. Reid IR, Gamble GD, Grey AB, Black DM, Ensrud KE, primary osteoblasts: confirmation. J Bone Miner Res. 1034–1043.
Browner WS, Bauer DC. beta-blocker use, BMD, and 2002;17(6):1135 author reply 1136. 128. Gat-Yablonski G, Shtaif B, Phillip M. Leptin stim-
fractures in the study of osteoporotic fractures. 112. Hess R, Pino AM, Rı́os S, Fernández M, Rodrı́guez JP. ulates parathyroid hormone related peptide ex-
J Bone Miner Res. 2005;20(4):613–618. High affinity leptin receptors are present in human pression in the endochondral growth plate. J Pediatr
99. Levasseur R, Dargent-Molina P, Sabatier JP, Marcelli mesenchymal stem cells (MSCs) derived from Endocrinol Metab. 2007;20(11):1215–1222.
C, Bréart G. Beta-blocker use, bone, mineral density, control and osteoporotic donors. J Cell Biochem. 129. Hamrick MW, Herberg S, Arounleut P, He HZ,
and fracture risk in older women: results from the 2005;94(1):50–57. Shiver A, Qi RQ, Zhou L, Isales CM, Mi Q-S. The
Epidemiologie de l’Osteoporose Prospective Study. 113. Sufang C, Nemoto K, Kurose R, Kobayashi N, Kubo adipokine leptin increases skeletal muscle mass and
J Am Geriatr Soc. 2005;53(3):550–552. K, Itabashi A. Different expression of leptin recep- significantly alters skeletal muscle miRNA expres-
100. Rejnmark L, Vestergaard P, Kassem M, Christoffersen tor variants in MC3T3-E1 osteoblastic cells and sion profile in aged mice. Biochem Biophys Res
BR, Kolthoff N, Brixen K, Mosekilde L. Fracture risk in ATDC-5 chondroblastic cells. J Bone Miner Res. 2000; Commun. 2010;400(3):379–383.
perimenopausal women treated with beta-blockers. 15(suppl 1):S466. 130. Liu C, Grossmann A, Bain S, Strachan M, Puemer D,
Calcif Tissue Int. 2004;75(5):365–372. 114. Matic I, Matthews BG, Wang X, Dyment NA, Bailey C, Humes J, Lenox J, Yamamoto G, Sprugel K,
101. Reid IR, Lucas J, Wattie D, Horne A, Bolland M, Worthley DL, Rowe DW, Grcevic D, Kalajzic I. Kuijper J, Weigle S, Dumam D, Moore E. Leptin
Gamble GD, Davidson JS, Grey AB. Effects of a Quiescent bone lining cells are a major source of stimulates cortical bone formation in obese (ob/ob)
beta-blocker on bone turnover in normal osteoblasts during adulthood. Stem Cells. 2016; mice. J Bone Miner Res. 1997;12(suppl 1):S115.
postmenopausal women: a randomized con- 34(12):2930–2942. 131. Gat-Yablonski G, Ben-Ari T, Shtaif B, Potievsky O,
trolled trial. J Clin Endocrinol Metab. 2005;90(9): 115. Gordeladze JO, Drevon CA, Syversen U, Reseland JE. Moran O, Eshet R, Maor G, Segev Y, Phillip M.
5212–5216. Leptin stimulates human osteoblastic cell pro- Leptin reverses the inhibitory effect of caloric re-
102. Wilding JP, Gilbey SG, Bailey CJ, Batt RA, Williams G, liferation, de novo collagen synthesis, and min- striction on longitudinal growth. Endocrinology.
Ghatei MA, Bloom SR. Increased neuropeptide-Y eralization: impact on differentiation markers, 2004;145(1):343–350.
messenger ribonucleic acid (mRNA) and decreased apoptosis, and osteoclastic signaling. J Cell Biochem. 132. Martin A, de Vittoris R, David V, Moraes R, Bégeot
neurotensin mRNA in the hypothalamus of the 2002;85(4):825–836. M, Lafage-Proust MH, Alexandre C, Vico L, Thomas

958 Reid et al Effects of Leptin on the Skeleton Endocrine Reviews, December 2018, 39(6):938–959
REVIEW

T. Leptin modulates both resorption and formation 140. Welt CK, Chan JL, Bullen J, Murphy R, Smith P, 148. Cornish J, Grey A, Callon KE, Naot D, Hill BL, Lin
while preventing disuse-induced bone loss in tail- DePaoli AM, Karalis A, Mantzoros CS. Recombinant CQX, Balchin LM, Reid IR. Shared pathways of
suspended female rats. Endocrinology. 2005;146(8): human leptin in women with hypothalamic osteoblast mitogenesis induced by amylin, adre-
3652–3659. amenorrhea. N Engl J Med. 2004;351(10):987–997. nomedullin, and IGF-1. Biochem Biophys Res Com-
133. Martin A, David V, Malaval L, Lafage-Proust MH, 141. Chou SH, Chamberland JP, Liu X, Matarese G, Gao mun. 2004;318(1):240–246.
Vico L, Thomas T. Opposite effects of leptin C, Stefanakis R, Brinkoetter MT, Gong H, Arampatzi 149. Cornish J, Callon KE, Bava U, Watson M, Xu X, Lin
on bone metabolism: a dose-dependent balance K, Mantzoros CS. Leptin is an effective treatment
JM, Chan VA, Grey AB, Naot D, Buchanan CM,
related to energy intake and insulin-like growth for hypothalamic amenorrhea. Proc Natl Acad Sci
Cooper GJS, Reid IR. Preptin, another peptide
factor–I pathway. Endocrinology. 2007;148(7): USA. 2011;108(16):6585–6590.
142. Conroy R, Girotra M, Shane E, McMahon DJ, product of the pancreatic beta-cell, is osteogenic
3419–3425.
Pavlovich KH, Leibel RL, Rosenbaum M, Korner J. in vitro and in vivo. Am J Physiol Endocrinol Metab.
134. Baek K, Bloomfield SA. Beta-adrenergic blockade
Leptin administration does not prevent the bone 2007;292(1):E117–E122.
and leptin replacement effectively mitigate

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/938/5088036 by Research 4 Life user on 22 November 2018


mineral metabolism changes induced by weight 150. Cornish J, Callon KE, Reid IR. Insulin increases his-
disuse bone loss. J Bone Miner Res. 2009;24(5):
loss. Metabolism. 2011;60(9):1222–1226. tomorphometric indices of bone formation in vivo.
792–799.
143. Sienkiewicz E, Magkos F, Aronis KN, Brinkoetter Calcif Tissue Int. 1996;59(6):492–495.
135. Bertoni L, Ferretti M, Cavani F, Zavatti M, Resca E,
M, Chamberland JP, Chou S, Arampatzi KM, Gao 151. Morroni M, De Matteis R, Palumbo C, Ferretti M,
Benelli A, Palumbo C. Leptin increases growth of
C, Koniaris A, Mantzoros CS. Long-term metre- Villa I, Rubinacci A, Cinti S, Marotti G. In vivo leptin
primary ossification centers in fetal mice. J Anat.
leptin treatment increases bone mineral density expression in cartilage and bone cells of growing
2009;215(5):577–583.
and content at the lumbar spine of lean hypo- rats and adult humans. J Anat. 2004;205(4):291–296.
136. Stunes AK, Westbroek I, Gordeladze JO, Gustafsson
leptinemic women. Metabolism. 2011;60(9):
BI, Reseland JE, Syversen U. Systemic leptin ad-
1211–1221.
ministration in supraphysiological doses maintains
144. Foo J-P, Polyzos SA, Anastasilakis AD, Chou S,
bone mineral density and mechanical strength Acknowledgments
Mantzoros CS. The effect of leptin replacement on
despite significant weight loss. Endocrinology. 2012; Financial Support: Supported by the Health Research
parathyroid hormone, RANKL-osteoprotegerin axis,
153(5):2245–2253. and Wnt inhibitors in young women with hypo-
Council of New Zealand.
137. Hamrick MW, Ding KH, Ponnala S, Ferrari SL, Isales Correspondence and Reprint Requests: Ian R. Reid,
thalamic amenorrhea. J Clin Endocrinol Metab. 2014;
CM. Caloric restriction decreases cortical bone MD, Faculty of Medical and Health Sciences, University of
99(11):E2252–E2258.
mass but spares trabecular bone in the mouse Auckland, Private Bag 92019, Auckland 1132, New Zealand.
145. Maness LM, Kastin AJ, Farrell CL, Banks WA. Fate
skeleton: implications for the regulation of bone E-mail: i.reid@auckland.ac.nz.
of leptin after intracerebroventricular injection
mass by body weight. J Bone Miner Res. 2008;23(6): Disclosure Summary: The authors have nothing to
into the mouse brain. Endocrinology. 1998;139(11):
870–878. disclose.
4556–4562.
138. Iwaniec UT, Dube MG, Boghossian S, Song H, 146. Trotter-Mayo RN, Roberts MR. Leptin acts in the
Helferich WG, Turner RT, Kalra SP. Body mass in- periphery to protect thymocytes from glucocorticoid- Abbreviations
fluences cortical bone mass independent of leptin mediated apoptosis in the absence of weight loss. BMD, bone mineral density; BMI, body mass index; CART,
signaling. Bone. 2009;44(3):404–412. Endocrinology. 2008;149(10):5209–5218. cocaine- and amphetamine-regulated transcript; CNS, cen-
139. Moran SA, Patten N, Young JR, Cochran E, Sebring N, 147. Otukonyong EE, Dube MG, Torto R, Kalra PS, Kalra tral nervous system; CTX, C-telopeptide of type I collagen;
Reynolds J, Premkumar A, Depaoli AM, Skarulis MC, SP. Central leptin differentially modulates ultradian FGF23, fibroblast growth factor-23; ICV, intracerebroventricular;
Oral EA, Gorden P. Changes in body composition in secretory patterns of insulin, leptin and ghrelin LepRb, long form of the leptin receptor; NPY, neuropeptide Y;
patients with severe lipodystrophy after leptin re- independent of effects on food intake and body SNS, sympathetic nervous system; Tph, tryptophan hydroxylase;
placement therapy. Metabolism. 2004;53(4):513–519. weight. Peptides. 2005;26(12):2559–2566. UCP1, uncoupling protein 1.

doi: 10.1210/er.2017-00226 https://academic.oup.com/edrv 959

Вам также может понравиться