13
Insulin Signaling in Osteoblasts Integrates Bone Remodeling and Energy Metabolism Mathieu Ferron, 1,5 Jianwen Wei, 1,5 Tatsuya Yoshizawa, 1,5 Andrea Del Fattore, 3 Ronald A. DePinho, 4 Anna Teti, 3 Patricia Ducy, 2 and Gerard Karsenty 1, * 1 Department of Genetics and Development 2 Department of Pathology and Cell Biology College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA 3 Department of Experimental Medicine, University of L’Aquila, L’Aquila, Italy 4 Department of Medicine and Genetics, Harvard Medical School, Boston, MA 02115, USA 5 These authors contributed equally to this work *Correspondence: [email protected] DOI 10.1016/j.cell.2010.06.003 SUMMARY The broad expression of the insulin receptor suggests that the spectrum of insulin function has not been fully described. A cell type expressing this receptor is the osteoblast, a bone-specific cell favoring glucose metabolism through a hormone, osteocalcin, that becomes active once uncarboxy- lated. We show here that insulin signaling in osteoblasts is necessary for whole-body glucose homeostasis because it increases osteocalcin activity. To achieve this function insulin signaling in osteoblasts takes advantage of the regulation of osteoclastic bone resorption exerted by osteoblasts. Indeed, since bone resorption occurs at a pH acidic enough to decarboxylate proteins, osteoclasts determine the carboxylation status and function of osteocalcin. Accordingly, increasing or decreasing insulin signaling in osteoblasts promotes or hampers glucose metabolism in a bone resorption-dependent manner in mice and humans. Hence, in a feed- forward loop, insulin signals in osteoblasts activate a hormone, osteocalcin, that promotes glucose metabolism. INTRODUCTION Bone is a multitasking tissue with mechanical, hematopoietic, and metabolic functions that result from the tight interplay between two bone-specific cell types, the osteoblast and the osteoclast. Bone also emerged recently as an endocrine organ regulating glucose metabolism (Fukumoto and Martin, 2009), a function that has been ascribed to date only to the osteoblast. The intricacy existing between osteoblasts and osteoclasts rai- ses the prospect, however, that the osteoclast may contribute to the endocrine role of the skeleton. Bone uses the osteoblast-specific secreted molecule osteo- calcin to favor glucose homeostasis. Circulating osteocalcin exists in two forms, carboxylated on 3 glutamate residues or undercarboxylated; the latter form being able to enhance insulin secretion by b-cells, insulin sensitivity and energy expenditure (Lee et al., 2007). Osteocalcin (Ocn), however, is not the only gene expressed in osteoblasts affecting glucose homeostasis. Esp, a gene encoding an intracellular tyrosine phosphatase called OST-PTP exerts, through its osteoblast expression, metabolic functions opposite to those of osteocalcin (Lee et al., 2007). Genetic and biochemical evidence show that Esp acts upstream of Ocn to inhibit its metabolic function. For instance, the meta- bolic phenotype of Esp / mice is fully corrected by removing one allele of Ocn even though Ocn +/ mice have no metabolic phenotype, and the fraction of undercarboxylated osteocalcin is significantly higher in Esp / than in wild-type (WT) mouse serum. The role of the osteoblast in regulating glucose metabolism revealed by these and other findings (Rached et al., 2010a; Yoshizawa et al., 2009) raises questions. The first one is to explain how OST-PTP, an intracellular tyrosine phosphatase, can influence the carboxylation and function of a secreted mole- cule like osteocalcin. A second issue is to provide evidence that the same bone-dependent regulation of glucose metabolism exists in humans since ESP is a pseudogene in this species (Cousin et al., 2004). A third question of physiological nature looming beyond these observations is whether insulin, in a feed- back loop, influences osteocalcin synthesis and/or activity. The insulin receptor is a tyrosine kinase whose activity must be tightly regulated since it can be activated in the absence of ligand (Kasuga et al., 1983). Receptor tyrosine kinases are often in- hibited by protein tyrosine phosphatases (PTPs) (Schlessinger, 2000) and PTP1B, which dephosphorylates the insulin receptor, is a major regulator of insulin signaling in hepatocytes and myo- cytes (Delibegovic et al., 2007, 2009). The fact that OST-PTP is a tyrosine phosphatase raises the testable hypothesis that the insulin receptor is one of its substrates. Our understanding of insulin signaling in various tissues has been profoundly altered by the analysis of mutant mouse strains 296 Cell 142, 296–308, July 23, 2010 ª2010 Elsevier Inc.

Insulin Signaling in Osteoblasts Integrates Bone ...2000) and PTP1B, which dephosphorylates the insulin receptor, is a major regulator of insulin signaling in hepatocytes and myo-

  • Upload
    others

  • View
    7

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Insulin Signaling in Osteoblasts Integrates Bone ...2000) and PTP1B, which dephosphorylates the insulin receptor, is a major regulator of insulin signaling in hepatocytes and myo-

Insulin Signaling in OsteoblastsIntegrates Bone Remodelingand Energy MetabolismMathieu Ferron,1,5 Jianwen Wei,1,5 Tatsuya Yoshizawa,1,5 Andrea Del Fattore,3 Ronald A. DePinho,4 Anna Teti,3

Patricia Ducy,2 and Gerard Karsenty1,*1Department of Genetics and Development2Department of Pathology and Cell Biology

College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA3Department of Experimental Medicine, University of L’Aquila, L’Aquila, Italy4Department of Medicine and Genetics, Harvard Medical School, Boston, MA 02115, USA5These authors contributed equally to this work*Correspondence: [email protected]

DOI 10.1016/j.cell.2010.06.003

SUMMARY

The broad expression of the insulin receptorsuggests that the spectrum of insulin function hasnot been fully described. A cell type expressingthis receptor is the osteoblast, a bone-specific cellfavoring glucose metabolism through a hormone,osteocalcin, that becomes active once uncarboxy-lated. We show here that insulin signaling inosteoblasts is necessary for whole-body glucosehomeostasis because it increases osteocalcinactivity. To achieve this function insulin signaling inosteoblasts takes advantage of the regulation ofosteoclastic bone resorption exerted by osteoblasts.Indeed, since bone resorption occurs at a pH acidicenough to decarboxylate proteins, osteoclastsdetermine the carboxylation status and function ofosteocalcin. Accordingly, increasing or decreasinginsulin signaling in osteoblasts promotes or hampersglucose metabolism in a bone resorption-dependentmanner in mice and humans. Hence, in a feed-forward loop, insulin signals in osteoblasts activatea hormone, osteocalcin, that promotes glucosemetabolism.

INTRODUCTION

Bone is a multitasking tissue with mechanical, hematopoietic,

and metabolic functions that result from the tight interplay

between two bone-specific cell types, the osteoblast and the

osteoclast. Bone also emerged recently as an endocrine organ

regulating glucose metabolism (Fukumoto and Martin, 2009),

a function that has been ascribed to date only to the osteoblast.

The intricacy existing between osteoblasts and osteoclasts rai-

ses the prospect, however, that the osteoclast may contribute

to the endocrine role of the skeleton.

296 Cell 142, 296–308, July 23, 2010 ª2010 Elsevier Inc.

Bone uses the osteoblast-specific secreted molecule osteo-

calcin to favor glucose homeostasis. Circulating osteocalcin

exists in two forms, carboxylated on 3 glutamate residues or

undercarboxylated; the latter form being able to enhance insulin

secretion by b-cells, insulin sensitivity and energy expenditure

(Lee et al., 2007). Osteocalcin (Ocn), however, is not the only

gene expressed in osteoblasts affecting glucose homeostasis.

Esp, a geneencodingan intracellular tyrosinephosphatasecalled

OST-PTP exerts, through its osteoblast expression, metabolic

functions opposite to those of osteocalcin (Lee et al., 2007).

Genetic and biochemical evidence show that Esp acts upstream

of Ocn to inhibit its metabolic function. For instance, the meta-

bolic phenotype of Esp�/� mice is fully corrected by removing

one allele of Ocn even though Ocn+/� mice have no metabolic

phenotype, and the fraction of undercarboxylated osteocalcin is

significantly higher inEsp�/� than inwild-type (WT)mouse serum.

The role of the osteoblast in regulating glucose metabolism

revealed by these and other findings (Rached et al., 2010a;

Yoshizawa et al., 2009) raises questions. The first one is to

explain how OST-PTP, an intracellular tyrosine phosphatase,

can influence the carboxylation and function of a secreted mole-

cule like osteocalcin. A second issue is to provide evidence that

the same bone-dependent regulation of glucose metabolism

exists in humans since ESP is a pseudogene in this species

(Cousin et al., 2004). A third question of physiological nature

looming beyond these observations is whether insulin, in a feed-

back loop, influences osteocalcin synthesis and/or activity.

The insulin receptor is a tyrosine kinasewhose activity must be

tightly regulated since it can be activated in the absence of ligand

(Kasuga et al., 1983). Receptor tyrosine kinases are often in-

hibited by protein tyrosine phosphatases (PTPs) (Schlessinger,

2000) and PTP1B, which dephosphorylates the insulin receptor,

is a major regulator of insulin signaling in hepatocytes and myo-

cytes (Delibegovic et al., 2007, 2009). The fact that OST-PTP is

a tyrosine phosphatase raises the testable hypothesis that the

insulin receptor is one of its substrates.

Our understanding of insulin signaling in various tissues has

been profoundly altered by the analysis of mutant mouse strains

Page 2: Insulin Signaling in Osteoblasts Integrates Bone ...2000) and PTP1B, which dephosphorylates the insulin receptor, is a major regulator of insulin signaling in hepatocytes and myo-

A

FE HG

B

50

50

3725

kDa GST GST-P

TP1B

WT

GST-P

TP1B

DA

Inpu

t (5%

)

Inpu

t (2%

)

100

75

kDa

100

75

10075

10075

10075

kDa

100

100

100

75

100

75100

75

75

75

kDa

100

75

100

75

10075

kDa

Coomassie staining

Coomassie staining

ROS17/2.8IB: InsR IB: InsR

OSBIB: InsR

IB: P-InsR(Y1150/Y1151)

IB: P-InsR(Y1150/Y1151)

pFLAGOST-WT-FLAGOST-DA-FLAG

IB: P-InsR(Y1150/Y1151)

IB: P-InsR(Y1146)

IB: InsRIB: InsR

IB: FLAG

IB: FLAG

IB: EphB2

IB: EphB2

15 min + EGF 15 min + Ins

Bone

IP: InsRIn vitro dephosphorylation

IP: F

LAG

Tota

l cel

l lys

ate

(2%

)

IB: P-InsR(Y1150/Y1151) IB: P-InsR

(Y1150/Y1151)

IB: P-InsR(Y1146)

IB: InsR

IB: InsR

IB: P-InsR(Y1150/Y1151)

IB: InsR

IB: P-EGFR(Y1068)

IB: EGFR

IP: InsRIn vitro dephosphorylation

GST-OSTPTP WT GST-PTP1B WT

Time (min.)20Esp-/-WT

105 6030 20 105 6030

Min. + Ins 1u/kg50 3015 12060

GST-OSTPTP DA-/+ NaVanadate (mM)

0.10 2.50.5 12.5

IB: P-FoxO1(S256)IB: FoxO1

GST

GST-P

TP1B

WT

GST-P

TP1B

DA

GST-O

STPT

P W

TGST

-OST

PTP

DA

GST-O

STPT

P W

T

GST-O

STPT

P DA

100

75

C D

Osteoblasts

Coomassie staining

WAT

kDa10075

10075

50

50

37

InsR

Live

r

Muscle

Calva

ria

OSB

β−Actin

Fem

ur

-- + -- ++- - +- --+ - -+ -

Figure 1. Insulin Receptor Is a Substrate of OST-PTP in Osteoblasts

(A) Western blot analysis of insulin receptor (InsR) expression in tissues and primary osteoblasts (OSB).

(B) In vivo phosphorylation of InsR and EGFR in bone following injection of a bolus of insulin.

(C) In vitro substrate trapping. Extracts from pervanadate-treated ROS17/2.8 cells or primary osteoblasts (OSB) were pulled down using GST or WT and DA

mutants of OST-PTP and PTP1B GST-fusion proteins. InsR was detected by western blot.

(D) In vitro substrate trapping conducted as described in (C) in absence or presence of increasing concentration of sodium orthovanadate (NaVanadate).

(E) In vivo substrate trapping. OST-PTP-WT and –DA FLAG tagged proteins were immunoprecipitated from ROS17/2.8 cells after 15 min stimulation with EGF

(100 ng/ml) or insulin (100 nM). Immunoprecipitated proteins (IP) and total cell lysates were then analyzed by western blot.

(F) In vitro dephosphorylation assay. Hyperphosphorylated InsR was immunoprecipitated (IP) from pervanadate-treated ROS17/2.8 extracts, incubated with

indicated recombinant proteins for 30 min and visualized by western blot.

(G) Time course of InsR dephosphorylation in vitro by OST-PTP and PTP1B. The experiment was conducted as in (F), except incubations were stopped at the

indicated times.

(H) Phosphorylation of InsR and FoxO1 in unstimulated WT and Esp�/� osteoblasts.

See also Figure S1.

lacking the insulin receptor in only one cell type (Bluher et al.,

2002; Bruning et al., 1998; Konner et al., 2007; Kulkarni et al.,

1999; Michael et al., 2000). Surprisingly, these studies failed to

demonstrate a major influence of insulin signaling in the control

of whole-body glucose homeostasis in two classical insulin

target tissues, muscle, and white fat (Bluher et al., 2002; Bruning

et al., 1998). An implication of these observations is that insulin

may act in additional organs in order tomaintain glucose homeo-

stasis. This hypothesis is consistent with the fact that the insulin

receptor is expressed inmany cell types where its functions have

not yet been analyzed. This is particularly relevant to the osteo-

blast since it expresses the insulin receptor and regulates insulin

secretion (Figure 1A) (Lee et al., 2007).

Here, we show that the insulin receptor is a substrate of

OST-PTP and PTP1B in mouse and human osteoblasts, respec-

tively. As a result, insulin signaling in osteoblasts enhances

osteocalcin activity and impacts glucose homeostasis by

promoting the ability of osteoblasts to enhance bone resorption.

Indeed, because the acid pH in the resorption lacuna allows

protein decarboxylation it is ultimately the activity of the osteo-

clast that determines the carboxylation status and function of

osteocalcin secreted by the osteoblast. These results reveal

a pH-dependent mechanism of activation for a hormone and

identify insulin signaling in osteoblasts as a critical link between

bone remodeling and energy metabolism.

RESULTS

The Insulin Receptor Is a Substrate of OST-PTPin Mouse OsteoblastsIn order to define the mechanism whereby Esp affects osteocal-

cin carboxylation, we asked whether osteocalcin and/or

enzymes required for its carboxylation, g-carboxylase and

Vkorc1 (Sadler, 2004), are phosphorylated on tyrosine residues

and could be substrates of OST-PTP. We failed to detect tyro-

sine phosphorylation of these enzymes or of an osteocalcin

peptide including all tyrosine residues of this molecule (Figures

S1A–S1C available online). We also failed to detect physical

interactions between osteocalcin, g-carboxylase, or Vkorc1,

and OST-PTP (Figures S1D–S1F). Although these negative

Cell 142, 296–308, July 23, 2010 ª2010 Elsevier Inc. 297

Page 3: Insulin Signaling in Osteoblasts Integrates Bone ...2000) and PTP1B, which dephosphorylates the insulin receptor, is a major regulator of insulin signaling in hepatocytes and myo-

results must be interpreted cautiously, they imply that OST-PTP

does not affect osteocalcin carboxylation by acting directly on

osteocalcin, g-carboxylase, or Vkorc1.

To pursue our search, we used substrate trapping, an assay

using phosphatase domains in which a single amino acid substi-

tution abrogates their catalytic activity, not their ability to recog-

nize substrates (Flint et al., 1997).

We used OST-PTP active phosphatase domain (Chengalvala

et al., 2001) to test whether the insulin receptor (InsR) was

a substrate of OST-PTP for the following reasons. First, dephos-

phorylation of the InsR by another tyrosine phosphatase, PTP1B,

regulates insulin signaling in other cell types (Delibegovic et al.,

2007; Delibegovic et al., 2009). Second, OST-PTP has homology

in its catalytic domain to phosphatases dephosphorylating the

InsR (Figure S1G). Third, the InsR is abundant in osteoblasts

and treatment of osteoblasts with insulin increases phosphoryla-

tion of the InsR and its downstream targets AKT, GSK3b, FoxO1

and p70S6K (Figure 1A and Figure S1H). Fourth, InsR phosphor-

ylation increased in bone in a time-dependent manner following

insulin injection in vivo (Figure 1B). Furthermore, we reasoned

that if osteoblasts favor glucose homeostasis insulin signaling

should influence osteoblast biology.

The InsR interacted with a mutated form of OST-PTP or of

PTP1B, a positive control, but not with negative controls such

as wild-type (WT) OST-PTP, PTP1B or glutathione S-transferase

(GST); InsR interaction with OST-PTP was competed by

increasing concentrations of orthovanadate, OST-PTP inter-

acted with InsR in cells following insulin treatment only, and

did not interact with an unrelated tyrosine kinase receptor

(Figures 1C–1E). Furthermore, WT OST-PTP dephosphorylated

in vitro InsR while a mutant form of OST-PTP lacking its phos-

phatase activity did not (Figures 1F and 1G). Consistent with

these results, phosphorylation of InsR on tyrosines 1150 and

1151 and of FoxO1, a target of insulin signaling (Puigserver

et al., 2003), was increased in Esp�/� osteoblasts (Figure 1H

and Figures S1I and S1J). These experiments provide congruent

biochemical evidence indicating that InsR is a substrate of OST-

PTP in mouse osteoblasts.

Insulin Signaling in Osteoblasts Is a Determinantof Whole-Body Glucose MetabolismThat insulin signaling in osteoblasts is regulated by OST-PTP,

a phosphatase affecting glucose metabolism, implies that it

also influences this process. To address this question, we gener-

ated osteoblast-specific Insulin Receptor (InsR)-deficient mice

(InsRosb�/� mice) by crossing a1(I)collagen-Cre transgenic

mice, which delete genes in osteoblasts only (Dacquin et al.,

2002), with mice harboring a floxed allele of InsR (Figures S2A–

S2C). Western blot analysis of osteoblasts showed at least

60%deletion of the InsR in osteoblasts but not in liver, pancreas,

white adipose tissue, muscle, and brain of InsRosb�/� mice

(Figures S2D–S2F).

InsRosb�/� mice were born at the expected Mendelian ratio

and at 8 weeks of age presented a significant elevation of blood

glucose in the fed state and a significant reduction of serum

insulin levels in the fasted or fed state, glucagon levels being

unaffected (Figures 2A and 2B and Figure S2G). A glucose-stim-

ulated insulin secretion (GSIS) test verified that insulin secretion

298 Cell 142, 296–308, July 23, 2010 ª2010 Elsevier Inc.

was decreased in InsRosb�/� mice (Figure 2C). There was also

a decrease in the number of islets, islet size, b-cell mass, b-cell

proliferation and insulin content in InsRosb�/� compared to

control pancreata (Figure 2D). As a result of this decrease in

insulin secretion a glucose tolerance test (GTT) demonstrated

glucose intolerance in InsRosb�/� mice (Figure 2E and

Figure S2H). Insulin tolerance, as measured by an insulin toler-

ance test (ITT), was also significantly reduced in InsRosb�/�

compared to control mice (Figure 2F and Figure S2I). Hence,

analysis of InsRosb�/� mice revealed that insulin signaling in

osteoblasts contributes to whole-body glucose homeostasis

by increasing b-cell proliferation and insulin secretion and energy

expenditure (Figure 2H and Figures S2J and S2K).

Insulin often transmits its signal by inhibiting FoxO1 activity.

Since FoxO1 affects glucose homeostasis through its expres-

sion in osteoblasts (Rached et al., 2010a), we asked whether it

was downstream of the insulin signaling pathway in these cells

by generating InsRosb�/� mice lacking one copy of FoxO1 only

in osteoblasts (InsRosb�/�;FoxO1osb

+/� mice). As would be

expected if FoxO1 does lie downstream of insulin signaling in

osteoblasts, decreasing its expression corrected the glucose

intolerance of InsRosb�/� mice. This result indicates that inhibit-

ing FoxO1 is a means whereby insulin signaling in osteoblasts

affects glucose homeostasis (Figures 2I–2L).

Insulin Signaling in Osteoblasts Acts Upstreamof Osteocalcin

Since OST-PTP inhibits osteocalcin activity, the observation that

InsR is its substrate implies that insulin signaling in osteoblasts

and osteocalcin lie in the same metabolic pathway.

To test this hypothesis, we first generated mice lacking one

allele of Osteocalcin and one allele of InsR in osteoblasts

(Ocn+/�;InsRosb+/� mice), hypothesizing that if the two genes

lie in the same pathway these compound heterozygous mutant

mice should display a phenotype reminiscent of the one

observed in Ocn�/� mice. Indeed, whether we studied glucose

in fed state, insulin secretion, glucose or insulin tolerance,

Ocn+/�;InsRosb+/� mice had metabolic abnormalities similar to

the ones of Ocn�/� mice (Figures 3A–3E), InsRosb+/� and

Ocn+/� mice were undistinguishable from control littermates.

Likewise, if OST-PTP inhibits the function of insulin signaling

in osteoblasts one would anticipate that the metabolic pheno-

type of Esp�/� mice should be corrected by decreasing insulin

signaling in osteoblasts. Accordingly, removing one allele of

InsR in osteoblasts from Esp�/� mice normalized glucose

and insulin tolerance (Figures 3F–3H). These two experiments

provide genetic evidence that a function of insulin signaling

in osteoblasts is to favor osteocalcin metabolic activity

(Figure 3I).

That reducing insulin signaling in osteoblasts corrected the

enhanced glucose and insulin tolerance of Esp�/� mice was

important in its own right, because it indicated that themetabolic

phenotype of Esp�/� mice is secondary to an increase in insulin

signaling in osteoblasts. This contention is supported by the

increased phosphorylation of the InsR in Esp�/� osteoblasts

(Figure 1H). Thus, in subsequent experiments studying insulin

signaling in osteoblasts, we relied on both loss- (InsRosb�/�)

and gain-of-function (Esp�/�) models.

Page 4: Insulin Signaling in Osteoblasts Integrates Bone ...2000) and PTP1B, which dephosphorylates the insulin receptor, is a major regulator of insulin signaling in hepatocytes and myo-

A

F

Islet # /mm2

Insulincontent (ng/μg)

β−cell area (%)

β−cellmass (mg)

Ki67 positive cells (%)

D

Blo

od G

luco

se (m

g/dl

)

400

300

200

100

Min. after glucose injection

InsRfl/fl (n=11) InsRosb-/- (n=12)

% o

f Ini

tial G

luco

se

100

80

60

Min. after insulin injection0 30 60 90 120

InsRfl/fl (n=5)

InsRosb-/-(n=5)

B C E

Blo

od G

luco

se (m

g/dl

)

120

100

140

160

180

0.5

1.0Se

rum

Insu

lin (n

g/m

l)

InsRfl/fl (n=10) InsRosb-/- (n=10)

InsRfl/fl (n=12) InsRosb-/- (n=14)

*

*

InsRfl/fl (n=21) InsRosb-/- (n=17)

InsRfl/fl (n=12) InsRosb-/- (n=6)

12.2 ± 0.9 8.4 ± 0.8**

****

**

0 30 60 120

**

**

**0.46 ± 0.04 0.39 ± 0.03*

0.44 ± 0.05 0.34 ± 0.02*

1.21 ± 0.12 0.98 ± 0.06*

*

Fasted Fed

0.73 ± 0.12 0.29 ± 0.04*Seru

m In

sulin

(ng/

ml) 3

2

1

Min. after glucose injection0 10 30

I J

Blo

od g

luco

se (m

g/dl

) 450

350

250

150

Min. after glucose injection

50 % o

f Ini

tial G

luco

se

50

100

75

Min. after insulin injection

InsRfl/fl (n=19)

InsRosb-/-;FoxO1osb+/- (n=12) InsRosb-/- (n=13)

InsRfl/fl (n=19)

InsRosb-/-;FoxO1osb+/- (n=10) InsRosb-/- (n=10)

Seru

m In

sulin

(ng/

ml)

2

1

H

InsRfl/fl (n=22)

InsRosb-/-;FoxO1osb+/- (n=12) InsRosb-/- (n=10)

Min. after glucose injection0 10 30 0 30 60 120

**

**

0 30 60 90 120

**

* * *

G InsRfl/fl (n=12) InsRosb-/- (n=12)

VO2

(ml/k

g/hr

)

3000

2600

3400

3800

4200

***

***

VCO

2 (m

l/kg/

hr)

3000

2600

3400

3800

4200

**

Day Night Day Night

HEA

T (k

cal/k

g/hr

)

16

10

18

20

22

**

Day Night

14

12

**

**

**

Are

a un

der t

he c

urve

10

20

30*

24000

28000

32000

*

7000

8000

9000

GSIS GTT ITT

Are

a un

der t

he c

urve

10

20

30

24000

28000

32000*

GSIS GTT ITT

InsRfl/fl InsRosb-/-

InsRfl/fl

InsRosb-/-;FoxO1osb+/- InsRosb-/-

**

*

7000

8000

9000

K L

***

**

**

**

**

**

**

**

****

*****

Figure 2. Decreased Insulin Secretion, Glucose Tolerance, and Insulin Tolerance in InsRosb�/� Mice

All experiments compare 8- to 12-week-old male mice. Genotypes are indicated in each panel.

(A) Random-fed blood glucose levels.

(B) Fasted and random-fed insulin levels.

(C) GSIS.

(D) Pancreas insulin content and histomorphometric comparisons of islet number, islet size, b-cell mass, and Ki67 immunoreactive cells in pancreatic islets.

(E) GTT.

(F) ITT.

(G) Area under the curve of (C), (E), and (F).

(H) Energy-balance data: oxygen consumed (VO2), carbon dioxide produced (VCO2), and heat.

(I–K) Rescue of the InsRosb�/� phenotype by FoxO1 haploinsufficiency, (I) GSIS, (J) GTT, (K) ITT.

(L) Area under the curve of (I)–(K).

*p < 0.05, **p < 0.01, and ***p < 0.001 versus InsRfl/fl in (A)–(H) (t test or ANOVA) or versus InsRfl/fl and InsRosb�/�;FoxO1osb

+/� in (I)–(L) (ANOVA).

See also Figure S2.

Insulin Signaling in Osteoblasts Inhibits OsteocalcinCarboxylationTo ascertain that insulin signaling in osteoblasts regulates

osteocalcin activity, we developed a dual ELISA quantifying

the ratio between undercarboxylated, i.e., active (GLU), and

carboxylated, i.e., inactive (GLA), circulating osteocalcin

(Figures S3A–S3E).

This ELISA detected an increase in undercarboxylated osteo-

calcin inEsp�/�mice,whichharbor an increase in insulin signaling

in osteoblasts (Figure 1H) but not in Esp�/�;InsRosb+/� mice that

do not (Figure 3J). In contrast, it showed that undercarboxylated

osteocalcin levels were decreased when insulin signaling is

impaired in osteoblasts (InsRosb�/� mice), even though Ocn

expression and serum levels were not affected (Figure 3J and

Figures S3F and S3G). A decrease in undercarboxylated osteo-

calcin was also observed in Ocn+/�;InsRosb+/� mice that display

a glucose intolerance similar to the one of the Ocn�/� and

InsRosb�/�mice (Figure3JandFigureS3H).Thesemeasurements

of undecarboxylated osteocalcin in variousmutantmouse strains

along with the genetic evidence presented above indicate that

insulin signaling in osteoblasts inhibits osteocalcin carboxylation,

i.e., is a positive regulator of its metabolic activity.

Insulin Signaling in Osteoblasts Favors Bone ResorptionIn a separate line of work, we noticed a marked decrease in the

serum level of CTx, a marker of bone resorption, in InsRosb�/�

Cell 142, 296–308, July 23, 2010 ª2010 Elsevier Inc. 299

Page 5: Insulin Signaling in Osteoblasts Integrates Bone ...2000) and PTP1B, which dephosphorylates the insulin receptor, is a major regulator of insulin signaling in hepatocytes and myo-

I

Blo

od G

luco

se (m

g/dl

) 400

300

200

100

0Min. after glucose injection

InsRfl/+ (n=13)Esp-/- (n=19)InsRosb+/- (n=13)Esp-/-; InsRosb+/- (n=8)

InsRfl/+ (n=13)Esp-/- (n=16)InsRosb+/- (n=11)Esp-/-; InsRosb+/- (n=8)

% o

f Ini

tial G

luco

se

100

80

60

40

Min. after insulin injection0

G

*

30 60 120

*

**

30 60 90 120

J

% o

f Ini

tial G

luco

se 100

80

60

40

Min. after insulin injection0 30 60 90

B

Blo

od G

luco

se (m

g/dl

) 400

300

200

100

0Min. after glucose injection

D

30 60 120

31 ± 225 ± 2*InsRosb-/- (n=7)

InsRfl/fl (n=9)

Esp-/- (n=6)Esp-/-;InsRosb+/- (n=6)

InsRfl/+ (n=8)

Ratio GLU/GLA (%)

15 ± 132 ± 2**20 ± 3

InsRosb+/- (n=5)Ocn+/- (n=6)

InsRfl/+ (n=7) 15 ± 313 ± 213 ± 3

Ocn+/-;InsRosb+/- (n=7) 8 ± 1°

A InsRfl/+ (n=5)InsRosb+/- (n=4)

Ocn+/-; InsRosb+/- (n=5)Ocn+/- (n=5)

Blo

od G

luco

se (m

g/dl

)

100

120

140

C

Seru

m In

sulin

(ng/

ml)

1.0

0.5

Min. after glucose injection0 10 30

1.5

F

**

120

160

180

FoxO1

P

InsulinReceptor

Esp

Osteoblast

Glu-OCN(active)

FoxO1P

??

Ocn-/- (n=5)

InsRfl/+ (n=17)InsRosb+/- (n=12)

Ocn+/-; InsRosb+/- (n=11)Ocn+/- (n=7)

Ocn-/- (n=8)

* #

InsRfl/+ (n=17) InsRosb+/- (n=16)

Ocn+/-; InsRosb+/- (n=11) Ocn+/- (n=7)

Ocn-/- (n=20)

InsRfl/+ (n=17) InsRosb+/- (n=12)

Ocn+/-; InsRosb+/- (n=11) Ocn+/- (n=7)

Ocn-/- (n=11)

**

*

#

#

Are

a un

der t

he c

urve

10

20

30

* ##

24000

28000

32000** #

8000

9000

10000*

##

GSIS GTT ITT

InsRfl/+ InsRosb+/-

Ocn+/-; InsRosb+/- Ocn+/-

Ocn-/-

E

Are

a un

der t

he c

urve

22000

24000

26000

28000

* ***

GTT ITT

InsRfl/+Esp-/- InsRosb+/- Esp-/-; InsRosb+/-

H

# #

8000

9000

10000

##

#

***

**

** #

** #

**#

#

* #

* # * # * #

*

**

**

****

**

Insulin

Figure 3. Genetic Interaction between InsR, Esp, and Osteocalcin

All experiments compare 6- to 8-week-old male mice unless otherwise noted.

(A) Random-fed blood glucose level.

(B) GSIS.

(C and F) GTT.

(D and G) ITT.

(E) Area under the curve of (B)–(D).

(H) Area under the curve of (F) and (G).

(I) Model of insulin signaling in osteoblasts.

(J) Carboxylation levels of serum osteocalcin assessed by GLU/GLA dual ELISA.

In (A)–(E), *p < 0.05 and **p < 0.01 InsRosb+/�;Ocn+/� versus InsRfl/+, InsRosb+/� and Ocn+/� (ANOVA). #p < 0.05 and ##p < 0.01 Ocn�/� versus InsRfl/+,

InsRosb+/� and Ocn+/� (ANOVA). In (F)–(H), *p < 0.05, **p < 0.01, and ***p < 0.001 versus InsRfl/+, InsRosb+/� and Esp�/�; InsRosb+/� (ANOVA). In (J),

*p < 0.05 versus InsRfl/fl (t test). �p < 0.05 versus InsRfl/+ (t test).

See also Figure S3.

mice, whereas this parameter was increased in Esp�/� mice

(Figure 4A). This data suggested that insulin signaling in osteo-

blasts favors bone resorption. Although not the focus of this study,

bone formationparameterswereseverelydecreased in InsRosb�/�

mice; as a result, these mice had a low bone mass (Figure S4A).

300 Cell 142, 296–308, July 23, 2010 ª2010 Elsevier Inc.

To prove that insulin signaling in osteoblasts affects bone

resorption, we used a coculture assay of osteoblasts and

osteoclasts (Takahashi et al., 1988). When cocultured with WT

osteoblasts, WT osteoclast precursor cells differentiated and

formed resorption pits. When WT osteoclast precursor cells

Page 6: Insulin Signaling in Osteoblasts Integrates Bone ...2000) and PTP1B, which dephosphorylates the insulin receptor, is a major regulator of insulin signaling in hepatocytes and myo-

F

H IG

C

2

1

Expr

essi

on re

lativ

eto

WT

(fold

)

Opg

0.5

1.0

Expr

essi

on re

lativ

eto

con

trol

s (fo

ld)

FoxO1fl/fl (n=4)FoxO1osb-/- (n=4)

Opg

2

1

Opg

leve

l rel

ativ

eto

Veh

icle

(fol

d)

D WT + Vehicle (n≥6)

InsR -/- + Vehicle (n≥4) InsR -/- + Ins 10 nM (n≥4)

WT + Ins 10 nM (n≥6)

mRNA

*

Expr

essi

on re

lativ

eto

pFL

AG

(fol

d)

pFLAG (n=3) FoxO1-FLAG (n=3)

2

1

Opg

*1.0

0.5

Opg

leve

l rel

ativ

eto

Con

siR

NA

(fold

)

Con siRNA (n≥3) FoxO1 siRNA (n≥4)

*

Seru

m C

Tx (n

g/m

l)

A

InsRfl/fl (n=13)

InsRosb-/-;FoxO1osb+/- (n=9)

InsRosb-/-;FoxO1osb+/- (n=7)

InsRobs-/- (n=11)

20

10

Seru

m C

Tx (n

g/m

l)

25

30

20

*

*

InsRfl/fl (n=11)InsRosb-/- (n=12)

Esp-/- (n=10)

35

40*

B

*

K

0.5 **

InsR fl/fl (n=7) InsR-/- (n=6)

Expr

essi

on re

lativ

eto

con

trol

s (fo

ld)

**

Ctsk Tcirg1

1

J

*

Expr

essi

on re

lativ

eto

con

trol

s (fo

ld)

*

Ctsk Tcirg1

2

Rankl Csf1

*

InsR fl/fl OSB (n=3)

Esp+/+ OSB (n=6) Esp-/- OSB (n=5)

InsR-/- OSB (n=3)

*

1

InsRfl/fl (n=11)InsRosb-/- (n=8)

Esp-/- (n=6)

*

*

6

4

2

Are

a co

vere

d by

re

sorp

tion

pits

(%)

*

*

InsR+/+ OSB (n=3) InsR-/- OSB (n=3)

Esp-/- OSB (n=3)Esp+/+ OSB (n=3)

Esp+/+ OSB Esp-/- OSB

InsR+/+ OSB InsR-/- OSB

L

0.5

*

InsRfl/fl (n=13) InsRosb-/- (n=12)

Opg

/Ran

kl ra

tio

1.2

0.8

300

200

100

0 **Ost

eocl

asts

num

ber

0 1.1 3.3 10 30 90OPG (ng / ml)

Are

a co

vere

d by

re

sorp

tion

pits

(%)

3

2

1

0

4

0 1.1 3.3 10 30 90**

***

OPG(ng / ml)0 3.3 30

OPG (ng / ml)

TRA

PPi

t

E

Trap

Secretion

* *

*****

mRNA Secretion

*

Esp+/+ OSB (n=5) Esp-/- OSB (n=5)

1.0

0.5

Opg

leve

l rel

ativ

eto

WT

(fold

)

Secretion

*

MEsp+/+ (n=10)

Esp+/+ (n=8)

300

200

100

0

n=3 n=3

*

**

Ost

eocl

asts

num

ber

Figure 4. Insulin Signaling in Osteoblasts

Favors Osteoclast Function

Mice genotypes are indicated in each panel.

(A) CTx serum levels in 8-week-old mice.

(B) Differentiation and resorptive ability of osteo-

clasts cocultured in presence of control or mutant

osteoblasts.

(C) Expression of Opg, Rankl, and Csf1 in mouse

osteoblasts by real-time PCR.

(D) Secretion of OPG by Esp�/� and WT osteo-

blasts.

(E) Opg expression and OPG secretion by osteo-

blasts following a 4 hr and 24 hr insulin treatment,

respectively.

(F) Dose-dependent effects of OPG on differentia-

tion and resorptive ability of osteoclasts in pres-

ence of 30 ng/ml RANKL and 10 ng/ml M-CSF.

Representative pictures of TRAP staining and pit

assay (left panel). Quantification of the number of

the pits area (middle panel) and of TRAP positive

cells (right panel).

(G and H) Real-time PCR analysis of Opg expres-

sion and/or secretion in ROS17/2.8 osteoblastic

cells transfected with empty (pFLAG) or FoxO1

expression (FoxO1-FLAG) vector (G) or in mouse

osteoblasts transfected with control siRNA (Con

siRNA) or FoxO1 siRNA (H).

(I and J) Expression of Opg in bone (I) and CTx

serum levels (J) in 1- and 3-month-old mice.

(K and L) Real-time PCR analysis of Ctsk, Tcirg1

and Trap expression in bone (K) and in osteoclasts

cocultured in presence of control or InsR�/� oste-

oblasts (L).

(M) Real-time PCR analysis of the Opg/Rankl

expression ratio in bones of 3-month-old male

mice.

In (A)–(D), (G)–(I), (K), and (L), *p< 0.05 and **p< 0.01

versus corresponding controls (t test). In (F), *p <

0.05 versus 0 ng/ml OPG (t test). In (E), *p < 0.05,

**p < 0.01, and ***p < 0.001 versus WT+vehicle

(ANOVA). In (J) and (M), *p < 0.05 versus InsRfl/fl

and InsRosb�/�;FoxO1osb+/� (ANOVA).

See also Figure S4.

were cocultured with InsR�/� osteoblasts, the area covered by

resorption pits was significantly decreased, whereas it increased

50% when cocultured with Esp�/� osteoblasts (Figure 4B). That

the number of osteoclasts was unaffected whether we used

Esp�/� or InsR�/� osteoblasts (Figure 4B) is consistent with the

notion that insulin signaling in osteoblasts promotes only the

function of osteoclasts.

We next studied expression of genes influencing bone

resorption in osteoblasts and noticed that expression of

Osteoprotegerin (Opg), a gene encoding a decoy receptor for

RANKL and a negative regulator of osteoclast function (Teitel-

baum and Ross, 2003), was increased 2-fold in InsR�/� and

Cell 142, 296–

decreased 50% in Esp�/� osteoblasts;

its secretion followed the same pattern

(Figures 4C–4E). That insulin treatment

of WT but not of InsR�/� osteoblasts

decreasedOpg expression and secretion

verified that it is an insulin target gene in

osteoblasts (Figure 4E). To demonstrate that a moderate

increase in OPG secretion inhibits the activity rather than the

differentiation of osteoclasts, we cultured osteoclast progenitor

cells in the presence of increasing amount of OPG (0–90 ng/ml).

Low amounts of OPG in the culture medium decreased the

surface covered by resorption pits but not the osteoclasts

number (Figure 4F).

Multiple evidences indicated that insulin signaling in osteo-

blasts favors bone resorption by inhibiting FoxO1. First, Opg

expression was increased 2-fold in ROS17/2.8 osteoblastic cells

overexpressing FoxO1 (Figure 4G). Second, siRNA-mediated

downregulation of FoxO1 decreased Opg expression and

308, July 23, 2010 ª2010 Elsevier Inc. 301

Page 7: Insulin Signaling in Osteoblasts Integrates Bone ...2000) and PTP1B, which dephosphorylates the insulin receptor, is a major regulator of insulin signaling in hepatocytes and myo-

secretion in mouse osteoblasts (Figure 4H and Figure S4B).

Third, the increase in bone resorption noted in FoxO1obs�/�

mice (Rached et al., 2010b) could be traced to a decrease in

Opg expression in bone (Figure 4I). Fourth, removing one allele

of FoxO1, in osteoblasts only, from InsRosb�/� mice normalized

bone resorption (Figure 4J).

Insulin Signaling in Osteoblasts Promotes theOsteoclasts Ability to Acidify the Bone ExtracellularMatrixNext, we sought to identify Opg-dependent molecular events

taking place in osteoclasts under the control of insulin signaling

in osteoblasts. Expression of CathepsinK (Ctsk) and Tcirg1,

two genes implicated in bone resorption, was decreased in

InsRosb�/� and increased in Esp�/� bones (Figure 4K). Ctsk

and Tcirg1 expression was also decreased in osteoclasts ob-

tained following coculture of WT osteoclast precursors cells

with InsR�/� osteoblasts (Figure 4L). For the rest of this study

we focused on Tcirg1, a gene encoding a vacuolar proton

pump subunit essential for acidification of the bone extracellular

matrix (ECM), precisely because acidification of the bone ECM is

a prerequisite for bone resorption (Teitelbaum and Ross, 2003).

Taken together, the biochemical, molecular, and genetic

evidence presented above indicates that insulin signaling in

osteoblasts decreases Opg expression and the Opg/Rankl ratio

(Figure 4M); this results in an increase in Tcirg1 expression, in

ECM acidification, and in bone resorption.

Bone Resorption as a Means to DecarboxylateOsteocalcinAn acid pH can decarboxylate proteins (Engelke et al., 1991),

and this raises the prospect that bone resorption, which occurs

at an acid pH, can decarboxylate osteocalcin in vivo.

To verify that an acid pH could decarboxylate and activate

osteocalcin, equal amounts of carboxylated osteocalcin were

incubated for 2 weeks at 37�C in solutions buffered at either

pH 7.5 or pH 4.5, the latter being the pH present in the resorption

lacunae (Silver et al., 1988). Each osteocalcin solution was then

analyzed by high-resolution mass spectrometry. This revealed

a peak at m/z 1325.103(+4) corresponding to a form of osteocal-

cin in which all three glutamic residues are carboxylated (molec-

ular weight: 5296.42 Da) and another peak at m/z 1314.108 (+4)

corresponding to an osteocalcin form in which one of the three

glutamic residues was not carboxylated (molecular weight:

5252.43 Da) (Figure 5A). The mass difference between these

two forms is 43.9 Da, which corresponds to one carboxylation

modification.

Importantly, the ratio of undercarboxylated to fully carboxyl-

ated osteocalcin was significantly increased when osteocalcin

was incubated at pH 4.5 (Figure 5A and Figure S5A). Tandem

mass spectrometry analysis of trypsin-digested samples

demonstrated that the presence of a GLU residue at position

13 was increased more than 2-fold upon incubation at pH 4.5

(Figure 5B), thus suggesting that this residue is highly suscep-

tible to decarboxylation. We also assessed themetabolic activity

of these two preparations of osteocalcin. When rat insulinoma

INS-1 cells were treated with carboxylated (pH 7.5) or undercar-

boxylated (pH 4.5) osteocalcin, only the latter form could

302 Cell 142, 296–308, July 23, 2010 ª2010 Elsevier Inc.

increase insulin secretion to the same extent as recombinant un-

carboxylated osteocalcin used as a positive control (Ferron et al.,

2008) (Figure 5C).

Given these results, we cultured osteoclast precursor cells in

the presence or absence of RANKL on bovine cortical bone

slides devitalized to exclude any endogenous osteoblastic

activity. Two days after osteoclasts were identifiable, total, car-

boxylated, and undercarboxylated osteocalcin were measured.

When osteoclast differentiation had been triggered by RANKL,

the levels of total and undercarboxylated (GLU) osteocalcin

were increased, while that of carboxylated (GLA) osteocalcin

was decreased. As a result, there was a 2-fold increase in the

GLU/GLA ratio (Figures 5D and 5E). Thus, the resorptive activity

of osteoclasts suffices to activate osteocalcin.

The Osteoclast Ability to Acidify the Bone ECM FavorsGlucose HomeostasisGiven the role of osteocalcin in glucose metabolism, we next

asked whether the osteoclasts ability to activate osteocalcin

affects glucose metabolism. For that purpose we analyzed

oc/oc mice harboring a loss-of-function mutation in Tcirg1 re-

sulting in osteopetrosis (Scimeca et al., 2000). The level of under-

carboxylated osteocalcin was decreased 30% in oc/oc

compared to WT serum (Figure 5F). Accordingly, conditioned

mediumof oc/oc calvaria cultures did not stimulate insulin secre-

tion by INS-1 cells (Figure 5G). Hence, in oc/ocmicewhose oste-

oclasts cannot acidify the bone ECM, circulating osteocalcin is

metabolically inactive.

Importantly, for our purpose, oc/oc mice were also glucose

intolerant (Figures 5H and 5I) with a marked decrease in serum

insulin levels, pancreas insulin content, and Insulin expression

in pancreas (Figures 5J–5L). To determine whether this pheno-

type was secondary to a function of Tcirg1 in osteoclasts, we

transplanted oc/oc fetal liver hematopoietic stem cells into WT

irradiated mice. Transplantation of mutant hematopoietic stem

cells resulted in high bonemass inWT recipient animals because

their osteoclasts could not form resorption pits (Figure 5M and

Figures S5B–S5F). Fasting glucose blood levels were increased

while serum insulin levels in fasted and fed states were

decreased in mice transplanted with oc/oc cells (Figures 5N

and 5O). A GSIS test demonstrated a defect in insulin secretion

in these mice, whereas a GTT showed glucose intolerance

(Figures 5P–5R). Energy expenditure was also decreased in

WT mice transplanted with oc/oc cells (Figure 5S and Figures

S5G and S5H). As expected, in mice transplanted with oc/oc

cells, there was a significant decrease in serum undercarboxy-

lated osteocalcin while the total osteocalcin level was

unchanged (Figure 5T and Figure S5I). These results demon-

strate that the osteoclasts ability to acidify the bone ECM is

both necessary and sufficient to activate osteocalcin and to

influence whole-body glucose metabolism.

Insulin Signaling in Osteoblasts Favors GlucoseHomeostasis in a Bone Resorption-Dependent MannerThe observations presented above beg the following question: Is

it because it favors bone resorption that insulin signaling in oste-

oblasts promotes glucose homeostasis?

Page 8: Insulin Signaling in Osteoblasts Integrates Bone ...2000) and PTP1B, which dephosphorylates the insulin receptor, is a major regulator of insulin signaling in hepatocytes and myo-

A

Inte

nsity

Inte

nsity

CB D1314.108+4

OC2GLA849.943+2

YLGASVPSPDPLEPTR

1325.103+4

OC3GLA

pH 7.5

Ratio (%)2GLA/3GLA

11 ± 0.3

89 ± 2.6***

RatioGLU13/GLA13

17 ± 0.1

42 ± 1***pH 4.5

m/z m/z Ocn (ng/ml)

10

5

Rat

io G

LU/G

LA (%

)

RANKL (n=3)Control (n=3)

*

871.937+2

YLGASVPSPDPLγPTR

pH 7.5

pH 4.5

5

15

Rat

io G

LU/G

LA (%

)

WT (n=5)oc/oc (n=5)

E

**

F

Insu

lin le

vel r

elat

ive

to m

ediu

m (f

old)

1.0

1.5

*

WT CM (n=11)oc/oc CM (n=11)

Medium (n=14) H

1.6

1.2

0.8

0.4

0Seru

m In

sulin

(ng/

ml)

WT (n=12)oc/oc (n=6)

*

10

G

Blo

od g

luco

se (m

g/dl

)

450

350

250

150

0 30 60 12050

WT (n=21)oc/oc (n=12)

WToc/oc

KI

Min. after glucose injection

WT (n=3)oc/oc (n=3)

****

* *0.2

0 Ins1 Ins2 Gcg

Pancreas

Expr

essi

on re

lativ

eto

WT

(fold

)

0.40.6

0.8

1.0

J

40

30

20

10

0

Panc

reas

Insu

lin

cont

ent (

ng/μ

g)

WT (n=12)oc/oc (n=6)

*

P

Blo

od g

luco

se (m

g/dl

)

400

300

200

100

0 30 60 120

WT+ WT (n=12)WT + oc/oc (n=12)

Min. after glucose injection

**

**

**Se

rum

Insu

lin (n

g/m

l) 1.0

0.5

1.5

O

Min. after glucose injection0 10 30

WT+ WT (n=9)WT + oc/oc (n=11)

WT+ WT WT + oc/oc

WT+ WT (n=8)WT + oc/oc (n=11)

WT+ WT (n=6)WT + oc/oc (n=7)

**

**

2

1

Are

a co

vere

d by

re

sorp

tion

pits

(%)

WT+ WT (n=3)WT + oc/oc (n=3)

Blo

od G

luco

se(m

g/dl

)

70

60

80

90

100

0.5

0.75

Seru

m In

sulin

(ng/

ml)

**

**

*

Fasted Fed

WT+ WT (n=12)WT + oc/oc (n=14)

WT+ WT (n≥8)WT + oc/oc (n≥7)

L

R

M N

0.25

00

25

Rat

io G

LU/G

LA (%

)

*20

15

5

10

Q

Insu

lin le

vel

rela

tive

to c

ontr

ol (f

old)

Bone extract (n=3)

RANKL (n=3)Control (n=3)Medium (n=3)

2

1

Ost

eoca

lcin

leve

l r

elat

ive

to c

ontr

ol (f

old) 3

Total GLA GLU

S T

**

*

Are

a un

der t

he c

urve

10

20

30

40*

20000

24000

28000

GSIS GTT

*

Are

a un

der t

he c

urve

20000

24000

28000

GTT

*

VO2

(ml/k

g/hr

)

3000

2600

3400

3800*

VCO

2 (m

l/kg/

hr)

2400

2000

2600

3000

3400

*

Day Night Day Night

HEA

T (k

cal/k

g/hr

)

16

10

18

20

22

*

Day Night

14

12

*

**

GluOC (n≥11)

GlaOC pH7.5 (n≥9)GlaOC pH4.5 (n≥9)

Control (n=15)

0.3 3

2 **********

1

******

**

**

**

Figure 5. An Acidic Environment Suffices to Decarboxylate Osteocalcin g-Carboxyglutamic Acid 13

(A) High-resolution mass spectrometry analysis of carboxylated osteocalcin incubated for 2 weeks at pH 7.5 or pH 4.5.

(B) Tandem mass spectrometry analysis of trypsin-digested samples of carboxylated osteocalcin incubated at pH 7.5 and pH 4.5.

(C) Insulin secretion from INS-1 cells following a 1 hr treatment with the indicated forms of osteocalcin.

(D) Total, GLA, and GLU osteocalcin released from bovine bone slices seeded with osteoclast progenitors treated with RANKL or vehicle or medium alone

measured by ELISA.

(E) GLU/GLA ratio of the osteocalcin released from bovine bone slices seeded with osteoclast progenitors treated with RANKL or vehicle and osteocalcin

chemically extracted from bone slide measured by dual ELISA.

(F–L) Analysis of 2- to 3-week-oldWT and oc/ocmice. (F) Carboxylation ratio of serum osteocalcin measured by dual ELISA. (G) Insulin secretion from INS-1 cells

following a 1 hr treatment with conditioned media (CM) from ex vivo cultures of calvaria from WT and oc/ocmice. (H) GTT at 2 weeks of age by IP injecting mice

wth glucose (1 g/kg) under random-fed condition. (I) Area under the curve of (H). (J) Random-fed insulin levels. (K) Pancreas insulin content. (L) Expression of

Insulin genes (Ins1 and Ins2) and Glucagon (Gcg) in pancreas by real-time PCR.

(M–T) Analysis of 10- to 12-week-oldWTmice transplanted with either WT (WT +WT) or oc/oc (WT + oc/oc) fetal liver hematopoietic stem cells. (M) Quantification

of the resorptive ability of osteoclasts derived from transplantedmice cultured in presence of RANKL andM-CSF. (N) Fasted blood glucose levels. (O) Fasted and

random-fed insulin levels. (P) GSIS. (Q) GTT. (R) Area under the curve of (P) and (Q). (S) Energy-balance data: oxygen consumed (VO2), carbon dioxide produced

(VCO2), and heat. (T) Carboxylation ratio of serum osteocalcin measured by dual ELISA.

In (C), **p < 0.01 and ***p < 0.001 versus control andGLA-OCNpH7.5 (ANOVA). In (D), (E), and (G), *p < 0.05 and **p < 0.01 versus control and/ormedium (ANOVA).

In (F), (I–O), and (R–T), *p < 0.05 and **p < 0.01 versus WT (t test). In (H), (P), and (Q), *p < 0.05 and **p < 0.01 versus WT (ANOVA).

See also Figure S5.

Cell 142, 296–308, July 23, 2010 ª2010 Elsevier Inc. 303

Page 9: Insulin Signaling in Osteoblasts Integrates Bone ...2000) and PTP1B, which dephosphorylates the insulin receptor, is a major regulator of insulin signaling in hepatocytes and myo-

To determine whether the influence of insulin signaling in oste-

oblasts on glucose metabolism depends on the osteoclasts

ability to acidify the bone ECM, we generated mice lacking

one allele of InsR in osteoblasts and harboring one oc allele

(InsRosb+/�;oc/+mice). In these mutant mice, osteocalcin under-

carboxylation was markedly decreased (Figure 6A); as a result

of this shift toward decreased osteocalcin activity, insulin secre-

tion, glucose, and insulin tolerance were impaired in InsRosb+/

�;oc/+ but not in InsRosb+/� or oc/+ mice (Figures 6B–6D and

Figure S6A). These data genetically link insulin signaling in oste-

oblasts, bone resorption, and whole-body glucose homeostasis.

If insulin signaling in osteoblasts promotes glucose homeo-

stasis in a bone resorption-dependent manner, then inhibiting

bone resorption in a model of increased insulin signaling in oste-

oblasts, such as the Esp�/� mice, should correct their metabolic

phenotypes. Indeed, and unlike Esp�/� mice, Esp�/�;oc/+ mice

had normal bone resorption, normal osteocalcin carboxylation

status (Figures 6E and 6F), normal insulin secretion, normal

glucose, and insulin tolerance (Figures 6G–6I and Figure S6B).

Likewise, when we treated Esp�/� mice with alendronate, an

inhibitor of osteoclast activity (Fisher et al., 1999), osteocalcin

carboxylation, insulin secretion, glucose, and insulin tolerance

were normalized (Figures 6J–6N and Figure S6C).

Lastly, if insulin signaling in osteoblasts promotes glucose

metabolism by increasing bone resorption, stimulating bone

resorption should rescue, at least partially, glucose intolerance

in WT mice. To test this contention, we fed WT mice a high-fat

diet and treated them with RANKL or GST as a negative control.

As expected, RANKL increased bone resorption parameters

(Figures S6D and S6E). This increase in bone resorption led to

a 3-fold increase in serum levels of undercarboxylated osteocal-

cin (Figure 6O). RANKL-treated mice fed a high-fat diet secreted

more insulin, were significantly less glucose intolerant, more

insulin tolerant and less fat than GST-treated mice (Figures 6P–

6S and Figure S6F). These three experiments demonstrate that

insulin signaling in osteoblasts cannot affect osteocalcin activity

and glucose metabolism if bone resorption is impaired.

Insulin Signaling in Human Osteoblasts, BoneResorption, Osteocalcin Activity, and GlucoseHomeostasisClinical studies indicate that osteocalcin is involved in glucose

homeostasis in humans (Hwang et al., 2009; Kanazawa et al.,

2009; Pittas et al., 2009). However, since ESP is a pseudogene

in humans (Cousin et al., 2004) osteocalcin activity must be regu-

lated by another phosphatase. The identification of InsR as a

substrate of OST-PTP provided a way to address this question.

We asked whether PTP1B, a tyrosine phosphatase able to

dephosphorylate INSR, was present in human osteoblasts.

PTP1B was markedly more abundant in human than in mouse

osteoblasts and decreasing its expression in human osteoblasts

increased INSR and FOXO1 phosphorylation and decreased

OPG expression (Figures 7A–7D). Conversely, siRNA-mediated

knockdown of INSR in human osteoblasts decreased FOXO1

phosphorylation and increased OPG expression (Figures 7D

and 7E). Finally, PTP1B could trap INSR in human osteoblasts

(Figure 7F). Thus, insulin signaling in human osteoblasts is regu-

lated by a tyrosine phosphatase and favors bone resorption.

304 Cell 142, 296–308, July 23, 2010 ª2010 Elsevier Inc.

The observations made in mice imply that patients harboring

a defect in bone resorption (osteopetrosis) should have low

serum undercarboxylated osteocalcin and serum insulin levels.

We tested this contention by analyzing patients with an auto-

somal dominant form of osteopetrosis. Three patients had

amissensemutation inClCN7, a gene required for acid secretion

in the resorption lacuna (Schaller et al., 2005); for the others,

although no genetic defect could be identified, a decrease of

acidification ability was demonstrated in osteoclasts cultured

ex vivo in presence of LysoSensor fluorescent pH indicators

(Figures S7A and S7B). Circulating undercarboxylated osteocal-

cin was significantly decreased in all patients, as were serum

insulin levels measured after feeding (Figure 7G). These data

support the notion that osteocalcin activity is determined in

humans, in part, by bone resorption, an aspect of bone remodel-

ing regulated by insulin signaling in osteoblasts.

DISCUSSION

This study demonstrates that insulin signaling in osteoblasts is

a significant determinant of whole-body glucose homeostasis.

Taking full advantage of the interplay between osteoblasts and

osteoclasts characterizing bone remodeling, insulin signaling in

osteoblasts achieves this function by favoring osteocalcin

decarboxylation (Figure 7H). Thus, this study identifies insulin

as a key molecular link between bone remodeling and energy

metabolism.

Insulin Signaling in Osteoblasts and GlucoseHomeostasisInsulin signaling and functions in various tissues have been

extensively studied in vivo in the last 10 years using mutant

mouse strains lacking the insulin receptor in only one cell type.

These studies have redefined the respective contributions of

various cell types to whole-body glucose homeostasis. For

instance, they showed that insulin signaling in b-cells is needed

for insulin secretion and, as expected, insulin signaling in

hepatocytes is needed for insulin sensitivity (Kulkarni et al.,

1999; Michael et al., 2000). In contrast, deletion of this receptor

in white adipose tissue (WAT) resulted in an improvement of

glucose metabolism (Bluher et al., 2002). Taken together these

various studies imply that insulin exerts yet to be identified

functions in other tissues, an idea congruous with the fact that

the insulin receptor is expressed in many cell types where its

function has not been studied yet.

In agreement with this hypothesis, we show here, through

the analysis of both loss- and gain-of-function models, that

insulin signaling in osteoblasts influences whole-body glucose

homeostasis by promoting insulin secretion. To the best of our

knowledge osteoblasts and hepatocytes are the only cell types

in which disrupting insulin signaling hampers glucose metabo-

lism in mice fed a normal diet.

Insulin Signaling in Osteoblasts, Osteocalcin Activation,and Bone ResorptionOne issue surrounding osteocalcin biology has been elucidating

how the intracellular phosphatase OST-PTP could favor carbox-

ylation. Here, we identify the insulin receptor as a bona fide

Page 10: Insulin Signaling in Osteoblasts Integrates Bone ...2000) and PTP1B, which dephosphorylates the insulin receptor, is a major regulator of insulin signaling in hepatocytes and myo-

J L M N

Blo

od g

luco

se (m

g/dl

)

450

350

250

150

0 30 60 120Min. after glucose injectionMin. after glucose injection50 %

of I

nitia

l Glu

cose

60

100

8070

Min. after insulin injection

Min. after glucose injectionMin. after glucose injection Min. after insulin injection

WT (n=17)Esp-/- (n=14)Esp-/-;oc/+ (n=16)

E G IH

****

*

*

**

****

**

WT + Veh. (n=10)

Esp-/- + Alend. (n=7)Esp-/- + Veh. (n=7)

Blo

od g

luco

se (m

g/dl

)

450

350

250

150

50 % o

f Ini

tial G

luco

se

25

100

75

50

0 30 60 90 120 Seru

m In

sulin

(ng/

ml) 2.0

1.5

1.0

0.5

0 10 300

Seru

m In

sulin

(ng/

ml) 2.0

1.5

1.0

0.5

0 10 300

Seru

m C

Tx (n

g/m

l)

*

Seru

m C

Tx (n

g/m

l)

10

20

30

50

40

10

20

30

40

WT + Veh. (n=15)

Esp-/- + Alend. (n=9)Esp-/- + Veh. (n=9)

WT + Veh. (n=8)

Esp-/- + Alend. (n=7)Esp-/- + Veh. (n=14)

WT + Veh. (n=8)

Esp-/- + Alend. (n=7)Esp-/- + Veh. (n=6)

WT (n=8)Esp-/- (n=7)Esp-/-;oc/+ (n=7)

F

*

Rat

io G

LU/G

LA (%

)

5

15

*

Rat

io G

LU/G

LA (%

)

WT + Veh. (n=8)

Esp-/- + Alend. (n=5)Esp-/- + Veh. (n=7)

K

P Q R

**

Seru

m In

sulin

(ng/

ml)

2.0

1.5

1.0

0.5

0 10 30Min. after glucose injection

GST (n=4)GST-RANKL (n=5)

**** ** ** * *

Blo

od g

luco

se (m

g/dl

)

450

350

250

150

Min. after glucose injection

50 % o

f Ini

tial G

luco

se

25

100

75

50

Min. after insulin injection0 60 120 180

Normal diet (n=3)HFD + GST (n=7)HFD + GST-RANKL (n=7)

10

40

Rat

io G

LU/G

LA (%

)

Normal diet (n=3)HFD + GST (n=8)HFD + GST-RANKL (n=9)

O

0 30 60 120

0 30 60 120

10

10

20

30

40

20

30

50

0 30 60 90 120

GST (n=9)GST-RANKL (n=8)

10

40Fa

t Pad

wei

ght o

ver

Bod

y w

eigh

t

S

20

30

50

*

*

Blo

od g

luco

se (m

g/dl

)

400

300

200

0 30 60 120Min. after glucose injectionMin. after glucose injection

100

% o

f Ini

tial G

luco

se

Min. after insulin injection

A B DC

Seru

m In

sulin

(ng/

ml) 1.2

0.8

0.4

0 10 300

**

*

*

°

Rat

io G

LU/G

LA (%

)

40

30

20

10 0 30 60 90 120

**

stop

Normal diet (n=7)HFD + GST (n=7)HFD + GST-RANKL (n=9)

WT (n=10)Esp-/- (n=18)Esp-/-;oc/+ (n=16)oc/+ (n=7)

WT (n=19)Esp-/- (n=16)Esp-/-;oc/+ (n=16)oc/+ (n=16)

WT (n=28)Esp-/- (n=13)Esp-/-;oc/+ (n=20)oc/+ (n=10)

50

90

InsRosb+/-;oc/+ (n=20)

InsRfl/+ (n=23) InsRosb+/- (n=11) InsRfl/+;oc/+ (n=18)

InsRosb+/-;oc/+ (n=26)

InsRfl/+ (n=19) InsRosb+/- (n=11) InsRfl/+;oc/+ (n=19)

InsRosb+/-;oc/+ (n=20)

InsRfl/+ (n=23) InsRosb+/- (n=13) InsRfl/+;oc/+ (n=18)

60

100

80

70

90

**

********

InsRosb+/-;oc/+ (n=12)

InsRfl/+ (n=8) InsRosb+/- (n=3) InsRfl/+;oc/+ (n=8)

**

*

*

* *

*

*

*

****

** **

**

** ** ** ***

**

**

** **

** **

*

* **

**

***

**

**

Figure 6. Insulin Signaling in Osteoblasts Favors Glucose Homeostasis by Promoting Bone Resorption

(A–I) Analysis of 7- to 9-week-old (A–D) or 6- to 7-week-old (E–I) male mice of indicated genotypes.

(J–N) Analysis of 6-week-old WT and Esp�/� mice treated with vehicle (Veh.) or alendronate (Alend., 80 mg/kg/week) for 4 weeks.

(O–S) Analysis of 16-week-old WT mice fed a normal or a high-fat diet (HFD) and treated with GST or GST-RANKL (0.8 mg/kg/day) for 8 weeks.

(A, F, K, and O) Carboxylation ratios of serum osteocalcin measured by dual ELISA.

(B, G, L, and P) GSIS.

(C, H, M, and Q) GTT.

(D, I, N, and R) ITT.

(E and J) CTx serum levels.

(S) Epididymal fat pad mass.

In (A), �p < 0.05 versus InsRfl/+ and InsRosb+/� (ANOVA). In (B)–(D), *p < 0.05 versus InsRfl/+, InsRosb+/�, and InsRfl/+;oc/+ (ANOVA). In (E)–(I), *p < 0.05

versus WT, Esp�/�;oc/+, and oc/+ (ANOVA). In (J)–(N), *p < 0.05 and **p < 0.01 versus WT and Esp�/� +Alend (ANOVA). In (O) and (S), *p < 0.05 and **p <

0.01 versus GST group (t test). In (P)–(R), *p < 0.05 and **p < 0.01, and ***p < 0.001 versus normal diet and GST groups (ANOVA).

See also Figure S6.

Cell 142, 296–308, July 23, 2010 ª2010 Elsevier Inc. 305

Page 11: Insulin Signaling in Osteoblasts Integrates Bone ...2000) and PTP1B, which dephosphorylates the insulin receptor, is a major regulator of insulin signaling in hepatocytes and myo-

A CB

GF

D E

0.5

1.0

Expr

essi

on re

lativ

eto

con

trol

(fol

d)

Con siRNA (n=3)

*

* INSR siRNA (n=3) PTP1B siRNA (n=3)

OPG CSF1

PTP1B

Mouse

OSB

Hum

an O

SBHu

man

OSB

+PTP

1B si

RNA

β−ACTIN

Con siRNATime (min.)post Ins (1μM)30

+ + + + + +

10 30 30 10 30

PTP1B siRNA

600 600

INSR siRNA

Con siRNA

P-FOXO1(S256)

P-FOXO1/O3a(T24/T32)

FOXO1

PTP1B

Time (min.)post Ins (50 nM)P-FOXO1(S256)

FOXO1

INSR

Age(Years)

Osteocalcinratio

GLU/GLA (%)

34 ± 7 20.2 ± 5.0

45 6.347 16.5

Controls(n=6-8)

ADO-1ADO-2

29 ± 8

41

Insulin(mU/l)

34 10.9ADO-3 14

64 10ADO-4 1635 9.9ADO-5 11

47 ± 16 20.1 ± 2.9 49 ± 18

Mal

esFe

mal

es

63 11.8ADO-6 17

Controls(n=4-9)

Vect

orPT

P1B

WT-

FLAG

PTP1

B DA

-FLA

GVe

ctor

PTP1

B W

T-FL

AGPT

P1B

DA-F

LAG

P-INSR(Y1150/Y1151)

EphB2

FLAG

IP: FLAG Total cell lysate(5%)

Ins (100 nM)

300 300

PTP1BsiRNA

Con siRNA

Time (min.)post Ins (5 nM)

PTP1B

P-INSR(Y1150/Y1151)

INSR

H

FoxO1

P

Gla-OCN(inactive)

InsulinReceptor

Esp

Osteoblast

Opg

Osteoclast

Gla-OCN(inactive)

Glu-OCN(active)

pH4.5Gla-OCN(inactive)

Bone extracellular matrix

Tcirg1 Glu-OCN(active)

Resorption lacuna

Insulin

Insulinsensitivetissuesβ-cells

FoxO1P

Esp

Ocn

Boneresorption

Figure 7. Insulin Signaling in Human Osteoblasts and Glucose Homeostasis

(A) Western blot analysis of PTP1B expression in human and mouse osteoblasts (OSB).

(B) Phosphorylation of INSR in human osteoblasts transfected with control (Con) or PTP1B siRNA.

(C) Phosphorylation of FOXO1 in human osteoblasts transfected with control (Con) or PTP1B siRNA.

(D) Real-time PCR analysis of OPG and CSF1 expression in human osteoblasts transfected with control siRNA (Con siRNA) or INSR siRNA or PTP1B siRNA.

(E) Phosphorylation of FOXO1 in human osteoblasts transfected with control (Con) or INSR siRNA.

(F) In vivo trapping of INSR by PTP1B DA mutant in human osteoblasts.

(G) Insulin levels (90 min postfeeding) and osteocalcin carboxylation ratio assessed by dual ELISA in human patients affected with dominant osteopetrosis (ADO)

and normal controls.

(H) Model of the bone resorption-dependent activation of osteocalcin by InsR. Insulin signaling in osteoblasts, which is inhibited by Esp, decreases in a

FoxO1-dependent manner OPG expression. This promotes bone resorption and in particular Tcigr1 expression and acidification of the bone ECM, which

promotes osteocalcin decarboxylation and as a result b-cells proliferation, insulin secretion, and insulin sensitivity.

See also Figure S7.

substrate of OST-PTP. This finding means that the metabolic

phenotype of the OST-PTP-deficient mice characterized by

improved whole-body glucose homeostasis (Lee et al., 2007) is

due to an increase in insulin signaling in osteoblasts.

More importantly, since OST-PTP regulates osteocalcin

function these results also placed de facto insulin signaling in

osteoblasts upstream of osteocalcin. To increase osteocalcin

bioactivity insulin signaling in osteoblasts acts in a ricocheting

manner using the huge amount of osteocalcin stored in the

bone ECM and the interplay between osteoblasts and osteo-

clasts. Specifically, it takes advantage of the ability of the

osteoblast to promote bone resorption. Since an acid pH is the

only known chemical condition allowing protein decarboxylation

(Engelke et al., 1991), bone resorption, which occurs at pH 4.5,

provides an ideal setting to decarboxylate and activate osteocal-

cin (Figure 7H).

The positive feedback loop existing between insulin signaling

in osteoblasts and osteocalcin functions implies that negative

regulators of this process may also exist. Leptin, another

hormone exerting a major influence on energy metabolism, is

one of them and does so also by modulating Esp expression

(Hinoi et al., 2008). Thus, these studies highlight the unexpected

importance of Esp in orchestrating the endocrine function of

bone.

306 Cell 142, 296–308, July 23, 2010 ª2010 Elsevier Inc.

Another issue about osteocalcin biology has been to provide

evidence that it has the same function in humans. Several argu-

ments indicate that it is the case. First, the tyrosine phosphatase

PTP1B fulfills in human osteoblasts the function that OST-PTP

fulfills in mouse osteoblasts. Second, insulin signaling in human

osteoblasts regulates OPG expression as it does in mouse

osteoblasts. Third, osteopetrotic patients and mice display the

same increase in osteocalcin carboxylation and decrease in

insulin blood levels. These results support a growing number of

observations linking osteocalcin or warfarin, a compound

decreasing its carboxylation, to the regulation of glucose

homeostasis in humans (Hwang et al., 2009; Kanazawa et al.,

2009; Pittas et al., 2009; Scheen, 2005). The notion that

hampering bone resorption has deleterious consequences on

glucose homeostasis is medically important since most drugs

used to treat osteoporosis target this aspect of bone remodeling.

Specifically, fasting plasma glucose levels are elevated in

osteoporotic women treated with drugs inhibiting bone resorp-

tion and serum undercarboxylated osteocalcin levels are

positively correlated with bone resorption in healthy women

(Kaji et al., 2009; Yamauchi et al., 2010).

Lastly, this work expands the relationship existing between

bone remodeling and energy metabolism (Karsenty, 2006) and

raises the testable hypothesis that the skeleton may exert

Page 12: Insulin Signaling in Osteoblasts Integrates Bone ...2000) and PTP1B, which dephosphorylates the insulin receptor, is a major regulator of insulin signaling in hepatocytes and myo-

additional endocrine influences on other, yet to be identified,

physiological functions.

EXPERIMENTAL PROCEDURES

Mice Generation

Generation of Esp�/� (C57BL/6J;129/Sv), Ocn�/� (C57BL/6J;129/Sv), and

FoxO1obs�/� (C57BL/6J;BALB/c) mice was reported (Dacquin et al., 2004;

Ducy et al., 1996; Rached et al., 2010a). InsRobs�/� mice were generated by

intercrossing the progeny of crosses between InsRflox/flox mice, that harbor

LoxP sites within introns 3 and 4 (Figure S2A), and a1(I)collagen-Cre transgenic

mice (Dacquin et al., 2002). oc/+ mice (C57BL/6J;C3H) were obtained

from The Jackson Laboratory. Genetic backgrounds of mice are as follows:

InsRosb-/- (C57BL/6J:87.5%; 129/Sv:12.5%), Ocn+/�;InsRosb+/� and Esp�/

�;InsRosb+/� (C57BL/6J;129/Sv), FoxO1osb

+/�;InsRosb�/� (C57BL/6J;BALB/c;

129/Sv), oc/+;InsRosb+/� (C57BL/6J;C3H;129/Sv). Control littermates were

used in all experiments. Mice genotypes were determined by PCR; primer

sequences are available upon request. CD45.2+ fetal liver stem cells isolated

from E14.5 WT or oc/oc embryos were transplanted (2 3 106/mice) via tail

vein injection into 5-week-old CD45.1+-irradiated WT recipient mice. Hemato-

poietic reconstitution was quantified by FACS analysis of blood cells collected

10 weeks posttransplantation (Figures S6B–S6E).

Metabolic Studies and Bioassays

Glucose tolerance test (GTT), glucose stimulated insulin secretion (GSIS),

insulin tolerance test (ITT), pancreas insulin content, histology, and high-fat

diet studies were performed as described (Lee et al., 2007). ELISA were

used tomeasuremouse/human insulin (Mercodia), plasma glucagon (ALPCO),

mouse CTx (RatLaps, IDS), bovine/human osteocalcin carboxylation ratios

(Takara), human TRAP (Bone TRAP Assay, IDS) and human CTx (Serum

Crosslaps, IDS).

Biochemistry and Gene Expression Studies

A complete description of all biochemical experiments is available as

Extended Experimental Procedures. All antibodies were obtained from Cell

Signaling Technology, with the exception of anti-PTP1B (R&D), anti-FLAG,

and anti-b-actin (Sigma). RNA isolation, cDNA preparation, and real-time

PCR analyses were carried out following standard protocols. Substrate-

trapping was conducted as described (Flint et al., 1997). Briefly, extracts

from pervanadate-treated cells were incubated in the presence of GST

recombinant protein and washed four times with lysis buffer. For in vitro

dephosphorylation assays, ROS17/2.8 cells were treated with pervanadate

(100 mM) for 30 min, and InsR was immunoprecipated. The immune complex

was then incubated at 30�C in the presence of 1 mg of recombinant GST,

GST-PTP, or GST-PTPDA for indicated times. Phosphorylation of InsR was

visualized by western blot. In in vivo trapping experiments, MG-63 or

ROS17/2.8 cells were transfected with FLAG-tagged expression vector, and

complexes were immunoprecipitated using anti-FLAG antibodies.

Cell Culture

Human osteoblastic cells hFOB 1.19, Saos-2 (ATCC) or mouse primary oste-

oblasts were transfected with siRNA pools (On-target, Dharmacon) according

to the manufacturer instructions. InsR+/+ and InsR�/� osteoblasts were

generated ex vivo by infecting InsRflox/flox with either GFP- or Cre-expressing

adenovirus (University of Iowa). Bone marrow progenitor cells and osteoblasts

cocultures were performed as previously described (Takahashi et al., 1988).

In vitro resorption activity of osteoclasts was measured using BD BioCoat

Osteologic Bone Cell Culture System.

Mass Spectrometry

Synthetic carboxylated osteocalcin (Bio-Synthesis Inc.) was incubated at

37�C in pH 7.5 or pH 4.5 0.1 M phosphate buffer for 2 weeks. Mass spectrom-

etry was run using Bruker Apex 9.4T Fourier Transform Ion Cyclotron

Resonance (FT-ICR) Mass Spectrometer and Daltonics DataAnalysis software

(v. 3.4) for analysis. Comparative quantification between carboxylated and

undercarboxylated osteocalcin were made based on exact mass measure-

ments and fit of isotopic peaks to that of theoretical isotopic patterns

(SNAP2 algorithm).

Statistics

Results are given as means ± standard errors of the mean except in human

studies in which standard deviations were used. Statistical analyses were

performed using unpaired, two-tailed Student’s t test for comparison between

two groups and One-way ANOVA test for more than two groups comparison.

For GTT, ITT, and GSIS, we performed two-way mixed designed ANOVA

and calculated Area Under the Curve (AUC) followed by student’s t test or

One-way ANOVA. For all experiments (*p or #p) % 0.05, (**p or ##p) % 0.01,

***p % 0.001.

SUPPLEMENTAL INFORMATION

Supplemental Information includes Extended Experimental Procedures,

Supplemental References, and seven figures and can be found with this article

online at doi:10.1016/j.cell.2010.06.003.

ACKNOWLEDGMENTS

We thank Drs. S. Kousteni, T. Zee, R.L. Levine, M. Tremblay, and J. Vacher for

reagents; Dr. J. Lacombe for help with transplantations; Drs. K. Henriksen and

J. Bollerslev for generously sharing patient serum samples; Dr. T. Lam at the

W.M. Keck Foundation Biotechnology Resource Laboratory, Yale University,

for FT-ICR mass spectral analyses; and Drs. J.K. Kim and F. Mauvais-Jarvis

for critical reading of the manuscript. This work was supported by a fellowship

from the Fond de la recherche en sante du Quebec (M.F.), grants from the

National Institutes of Health (G.K.), and the Juvenile Diabetes Research

Foundation (P.D.). G.K. is a founder and member of the scientific advisory

board of Escoublac Inc. P.D. is a founder and consultant of Escoublac Inc.

Received: January 4, 2010

Revised: March 29, 2010

Accepted: May 25, 2010

Published: July 22, 2010

REFERENCES

Bluher, M., Michael, M.D., Peroni, O.D., Ueki, K., Carter, N., Kahn, B.B., and

Kahn, C.R. (2002). Adipose tissue selective insulin receptor knockout protects

against obesity and obesity-related glucose intolerance. Dev. Cell 3, 25–38.

Bruning, J.C., Michael, M.D., Winnay, J.N., Hayashi, T., Horsch, D., Accili, D.,

Goodyear, L.J., and Kahn, C.R. (1998). A muscle-specific insulin receptor

knockout exhibits features of the metabolic syndrome of NIDDM without

altering glucose tolerance. Mol. Cell 2, 559–569.

Chengalvala, M.V., Bapat, A.R., Hurlburt, W.W., Kostek, B., Gonder, D.S.,

Mastroeni, R.A., and Frail, D.E. (2001). Biochemical characterization of

osteo-testicular protein tyrosine phosphatase and its functional significance

in rat primary osteoblasts. Biochemistry 40, 814–821.

Cousin,W., Courseaux, A., Ladoux, A., Dani, C., and Peraldi, P. (2004). Cloning

of hOST-PTP: the only example of a protein-tyrosine-phosphatase the

function of which has been lost between rodent and human. Biochem.

Biophys. Res. Commun. 321, 259–265.

Dacquin, R., Mee, P.J., Kawaguchi, J., Olmsted-Davis, E.A., Gallagher, J.A.,

Nichols, J., Lee, K., Karsenty, G., and Smith, A. (2004). Knock-in of nuclear

localised beta-galactosidase reveals that the tyrosine phosphatase Ptprv is

specifically expressed in cells of the bone collar. Dev. Dyn. 229, 826–834.

Dacquin, R., Starbuck, M., Schinke, T., and Karsenty, G. (2002). Mouse alpha1

(I)-collagen promoter is the best known promoter to drive efficient Cre recom-

binase expression in osteoblast. Dev. Dyn. 224, 245–251.

Delibegovic, M., Bence, K.K., Mody, N., Hong, E.G., Ko, H.J., Kim, J.K., Kahn,

B.B., and Neel, B.G. (2007). Improved glucose homeostasis in mice with

muscle-specific deletion of protein-tyrosine phosphatase 1B. Mol. Cell. Biol.

27, 7727–7734.

Cell 142, 296–308, July 23, 2010 ª2010 Elsevier Inc. 307

Page 13: Insulin Signaling in Osteoblasts Integrates Bone ...2000) and PTP1B, which dephosphorylates the insulin receptor, is a major regulator of insulin signaling in hepatocytes and myo-

Delibegovic, M., Zimmer, D., Kauffman, C., Rak, K., Hong, E.G., Cho, Y.R.,

Kim, J.K., Kahn, B.B., Neel, B.G., and Bence, K.K. (2009). Liver-specific

deletion of protein-tyrosine phosphatase 1B (PTP1B) improves metabolic

syndrome and attenuates diet-induced endoplasmic reticulum stress.

Diabetes 58, 590–599.

Ducy, P., Desbois, C., Boyce, B., Pinero, G., Story, B., Dunstan, C., Smith, E.,

Bonadio, J., Goldstein, S., Gundberg, C., et al. (1996). Increased bone

formation in osteocalcin-deficient mice. Nature 382, 448–452.

Engelke, J.A., Hale, J.E., Suttie, J.W., and Price, P.A. (1991). Vitamin K-depen-

dent carboxylase: utilization of decarboxylated bone Gla protein and matrix

Gla protein as substrates. Biochim. Biophys. Acta 1078, 31–34.

Ferron, M., Hinoi, E., Karsenty, G., and Ducy, P. (2008). Osteocalcin

differentially regulates beta cell and adipocyte gene expression and affects

the development of metabolic diseases in wild-type mice. Proc. Natl. Acad.

Sci. USA 105, 5266–5270.

Fisher, J.E., Rogers, M.J., Halasy, J.M., Luckman, S.P., Hughes, D.E.,

Masarachia, P.J., Wesolowski, G., Russell, R.G., Rodan, G.A., and Reszka,

A.A. (1999). Alendronate mechanism of action: geranylgeraniol, an interme-

diate in the mevalonate pathway, prevents inhibition of osteoclast formation,

bone resorption, and kinase activation in vitro. Proc. Natl. Acad. Sci. USA

96, 133–138.

Flint, A.J., Tiganis, T., Barford, D., and Tonks, N.K. (1997). Development of

‘‘substrate-trapping’’ mutants to identify physiological substrates of protein

tyrosine phosphatases. Proc. Natl. Acad. Sci. USA 94, 1680–1685.

Fukumoto, S., and Martin, T.J. (2009). Bone as an endocrine organ. Trends

Endocrinol. Metab. 20, 230–236.

Hinoi, E., Gao, N., Jung, D.Y., Yadav, V., Yoshizawa, T., Myers, M.G., Jr.,

Chua, S.C., Jr., Kim, J.K., Kaestner, K.H., and Karsenty, G. (2008). The

sympathetic tone mediates leptin’s inhibition of insulin secretion by

modulating osteocalcin bioactivity. J. Cell Biol. 183, 1235–1242.

Hwang, Y.C., Jeong, I.K., Ahn, K.J., and Chung, H.Y. (2009). The uncarboxy-

lated form of osteocalcin is associated with improved glucose tolerance and

enhanced beta-cell function in middle-aged male subjects. Diabetes Metab.

Res. Rev. 25, 768–772.

Kaji, H., Hisa, I., Inoue, Y., Naito, J., Sugimoto, T., and Kasuga, M. (2009).

Analysis of factors affecting increase in bone mineral density at lumbar spine

by bisphosphonate treatment in postmenopausal osteoporosis. J. Bone

Miner. Metab. 27, 76–82.

Kanazawa, I., Yamaguchi, T., Yamamoto, M., Yamauchi, M., Kurioka, S.,

Yano, S., and Sugimoto, T. (2009). Serum osteocalcin level is associated

with glucose metabolism and atherosclerosis parameters in type 2 diabetes

mellitus. J. Clin. Endocrinol. Metab. 94, 45–49.

Karsenty, G. (2006). Convergence between bone and energy homeostases:

Leptin regulation of bone mass. Cell Metab. 4, 341–348.

Kasuga, M., Fujita-Yamaguchi, Y., Blithe, D.L., White, M.F., and Kahn, C.R.

(1983). Characterization of the insulin receptor kinase purified from human

placental membranes. J. Biol. Chem. 258, 10973–10980.

Konner, A.C., Janoschek, R., Plum, L., Jordan, S.D., Rother, E., Ma, X., Xu, C.,

Enriori, P., Hampel, B., Barsh, G.S., et al. (2007). Insulin Action in AgRP-

Expressing Neurons Is Required for Suppression of Hepatic Glucose Produc-

tion. Cell Metab. 5, 438–449.

Kulkarni, R.N., Bruning, J.C., Winnay, J.N., Postic, C., Magnuson, M.A., and

Kahn, C.R. (1999). Tissue-specific knockout of the insulin receptor in

308 Cell 142, 296–308, July 23, 2010 ª2010 Elsevier Inc.

pancreatic beta cells creates an insulin secretory defect similar to that in

type 2 diabetes. Cell 96, 329–339.

Lee, N.K., Sowa, H., Hinoi, E., Ferron, M., Ahn, J.D., Confavreux, C., Dacquin,

R., Mee, P.J., McKee, M.D., Jung, D.Y., et al. (2007). Endocrine regulation of

energy metabolism by the skeleton. Cell 130, 456–469.

Michael, M.D., Kulkarni, R.N., Postic, C., Previs, S.F., Shulman, G.I.,

Magnuson, M.A., and Kahn, C.R. (2000). Loss of insulin signaling in hepato-

cytes leads to severe insulin resistance and progressive hepatic dysfunction.

Mol. Cell 6, 87–97.

Pittas, A.G., Harris, S.S., Eliades, M., Stark, P., and Dawson-Hughes, B.

(2009). Association between serum osteocalcin and markers of metabolic

phenotype. J. Clin. Endocrinol. Metab. 94, 827–832.

Puigserver, P., Rhee, J., Donovan, J., Walkey, C.J., Yoon, J.C., Oriente, F.,

Kitamura, Y., Altomonte, J., Dong, H., Accili, D., et al. (2003). Insulin-regulated

hepatic gluconeogenesis through FOXO1-PGC-1alpha interaction. Nature

423, 550–555.

Rached, M.-T., Kode, A., Silva, B.C., Jung, D.Y., Gray, S., Ong, H., Paik, J.-H.,

DePinho, R.A., Kim, J.K., Karsenty, G., et al. (2010a). FoxO1 expression in

osteoblasts regulates glucose homeostasis through regulation of osteocalcin

in mice. J. Clin. Invest. 120, 357–368.

Rached, M.-T., Kode, A., Xu, L., Yoshisawa, Y., Paik, J.-H., DePinho, R.A., and

Kousteni, S. (2010b). FoxO1 is a Positive Regulator of Bone Formation by

Favoring Protein Synthesis and Resistance to Oxidative Stress in Osteoblasts.

Cell Metab. 11, 147–160.

Sadler, J.E. (2004). Medicine: K is for koagulation. Nature 427, 493–494.

Schaller, S., Henriksen, K., Sorensen, M.G., and Karsdal, M.A. (2005). The role

of chloride channels in osteoclasts: ClC-7 as a target for osteoporosis

treatment. Drug News Perspect. 18, 489–495.

Scheen, A.J. (2005). Drug interactions of clinical importance with antihypergly-

caemic agents: an update. Drug Saf. 28, 601–631.

Schlessinger, J. (2000). Cell signaling by receptor tyrosine kinases. Cell 103,

211–225.

Scimeca, J.C., Franchi, A., Trojani, C., Parrinello, H., Grosgeorge, J., Robert,

C., Jaillon, O., Poirier, C., Gaudray, P., and Carle, G.F. (2000). The gene

encoding the mouse homologue of the human osteoclast-specific 116-kDa

V-ATPase subunit bears a deletion in osteosclerotic (oc/oc) mutants. Bone

26, 207–213.

Silver, I.A., Murrills, R.J., and Etherington, D.J. (1988). Microelectrode

studies on the acid microenvironment beneath adherent macrophages and

osteoclasts. Exp. Cell Res. 175, 266–276.

Takahashi, N., Akatsu, T., Udagawa, N., Sasaki, T., Yamaguchi, A., Moseley,

J.M., Martin, T.J., and Suda, T. (1988). Osteoblastic cells are involved in

osteoclast formation. Endocrinology 123, 2600–2602.

Teitelbaum, S.L., and Ross, F.P. (2003). Genetic regulation of osteoclast

development and function. Nat. Rev. Genet. 4, 638–649.

Yamauchi, M., Yamaguchi, T., Nawata, K., Takaoka, S., and Sugimoto, T.

(2010). Relationships between undercarboxylated osteocalcin and vitamin K

intakes, bone turnover, and bone mineral density in healthy women. Clin.

Nutr., in press, Published online March 23, 2010. 10.1016/j.clnu.2010.02.010.

Yoshizawa, T., Hinoi, E., Jung, D.Y., Kajimura, D., Ferron, M., Seo, J., Graff,

J.M., Kim, J.K., and Karsenty, G. (2009). The transcription factor ATF4 regu-

lates glucose metabolism in mice through its expression in osteoblasts. J.

Clin. Invest. 119, 2807–2817.