5
LETTERS Leptin is an adipocyte-derived hormone that plays a key role in energy homeostasis, yet resistance to leptin is a feature of most cases of obesity in humans and rodents. In vitro analysis suggested that the suppressor of cytokine signaling-3 (Socs3) is a negative-feedback regulator of leptin signaling involved in leptin resistance. To determine the functional significance of Socs3 in vivo, we generated neural cellspecific SOCS3 conditional knockout mice using the Cre–loxP system. Compared to their wild-type littermates, Socs3-deficient mice showed enhanced leptin-induced hypothalamic Stat3 tyrosine phosphorylation as well as pro-opiomelanocortin (POMC) induction, and this resulted in a greater body weight loss and suppression of food intake. Moreover, the Socs3-deficient mice were resistant to high fat dietinduced weight gain and hyperleptinemia, and insulin-sensitivity was retained. These data indicate that Socs3 is a key regulator of diet-induced leptin as well as insulin resistance. Our study demonstrates the negative regulatory role of Socs3 in leptin signaling in vivo, and thus suppression of Socs3 in the brain is a potential therapy for leptin-resistance in obesity. It was initially proposed that leptin has a primary role as an anti- obesity hormone; however, high concentrations of serum leptin levels are often associated with obesity, suggesting the presence of leptin resistance 13 . The leptin receptor is highly expressed in the hypothala- mus and belongs to the cytokine-receptor superfamily that activates the Janus kinasesignal transducers and activators of transcription (Jak-Stat) pathway 4 . Stat3 is apparently essential for leptin regulation of energy balance 57 . Several mechanisms for leptin resistance have been documented, including impaired leptin transport across the blood-brain barrier 8 and the presence of negative regulators of leptin signaling, such as the tyrosine phosphatases SH2-containing protein tyrosine phospatase-2 (SHP-2) (ref. 9) and protein-tyrosine phos- phatase 1B (PTP1B) 1012 as well as the suppressors of cytokine signal- ing family of molecules 13 . Peripheral leptin administration to mice rapidly induces Socs3 mRNA in hypothalamic regions 14 . Socs3 binds to the phosphorylated leptin receptor through its Src homology-2 (SH2) domain and inhibits Jak tyrosine kinase activity through its N-terminal kinase inhibitory region, which functions as a pseudo- substrate 13,15,16 . Furthermore, the C-terminal Socs box recruits the ubiquitin-transferase system and may mediate the degradation of receptor-Jak complexes 17 . Thus, Socs3 may be an important regulator of leptin signaling and could be involved in leptin-resistance. However, because these conclusions were based on the analysis of cell lines, the physiological significance of Socs3 in leptin signaling and its impact on obesity remained an open question. Because Socs3-deficient mice die during embryonic development as a result of placental defects 18 , a conditional knockout approach was used to determine the tissue-specific roles of Socs3 (ref. 19; Fig. 1a). To generate a neural cellspecific deletion, cre transgenic mice, in which Cre recombinase is expressed specifically in neural precursor cells under the nestin promoter-enhancer (cre nes ) 20 or synapsin promoter (cre syn ) 21 , were crossed with Socs3 flox/flox mice. Genomic DNA extracted from the brain was analyzed with PCR to verify deletion of Socs3. A 250base pair band corresponding to the deleted Socs3 allele (Socs3 del ) appeared in the PCR analysis of brain DNA from cre nes Socs3 flox/flox (NesKO) mice and cre syn Socs3 flox/flox (SynKO) mice but not in the DNA from control Socs3 flox/flox (wild type) mice (Fig. 1b). In these two types of conditional knockout mice, we con- firmed Socs3 deletion in the hypothalamus and cerebrum, but not in other tissues including heart, liver, kidney muscle and fat (Supplementary Fig. 1 online). RT-PCR analysis also indicated that Socs3 expression was greatly reduced in the brain but not in the other tissues examined (Supplementary Fig. 1 online). After leptin administration, Socs3 is exclusively expressed in the arcuate nucleus (ARC) of the hypothalamus, which coexpress as leptin receptors and neuropeptides that are considered to be essential in the regulation of food intake 14 . Consistent with previous reports, our real time RT-PCR analysis revealed that leptin treatment led to a twofold increase in Socs3 mRNA in wild-type mice (Fig. 1c). However, expres- sion of Socs3 mRNA in NesKO and SynKO mice was much lower than that in wild-type mice, irrespective of leptin administration (Fig. 1c). Immunohistological analysis with an antibody to Socs3 confirmed a marked induction of Socs3 protein in the ARC of the brain in 1 Division of Molecular and Cellular Immunology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan. 2 Department of Molecular Genetics, Institute of Life Science, Kurume University, 2432-3 Aikawa-machi, Kurume 839-0861, Japan. 3 Institute of Molecular Medicine and Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093-0641, USA. 4 Cardiovascular Research Institute and The Third Department of Internal Medicine, Kurume University, 67 Asahi-machi, Kurume 830-0011, Japan. Correspondence should be addressed to A. Y. ([email protected]). Published online 20 June 2004; doi:10.1038/nm1071 Socs3 deficiency in the brain elevates leptin sensitivity and confers resistance to diet-induced obesity Hiroyuki Mori 1 , Reiko Hanada 2 , Toshikatsu Hanada 1 , Daisuke Aki 1 , Ryuichi Mashima 1 , Hitomi Nishinakamura 1 , Takehiro Torisu 1 , Kenneth R Chien 3 , Hideo Yasukawa 4 & Akihiko Yoshimura 1 NATURE MEDICINE VOLUME 10 | NUMBER 7 | JULY 2004 739 © 2004 Nature Publishing Group http://www.nature.com/naturemedicine

Socs3 deficiency in the brain elevates leptin sensitivity and confers resistance to diet-induced obesity

  • Upload
    akihiko

  • View
    213

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Socs3 deficiency in the brain elevates leptin sensitivity and confers resistance to diet-induced obesity

L E T T E R S

Leptin is an adipocyte-derived hormone that plays a key role inenergy homeostasis, yet resistance to leptin is a feature ofmost cases of obesity in humans and rodents. In vitro analysissuggested that the suppressor of cytokine signaling-3 (Socs3)is a negative-feedback regulator of leptin signaling involved inleptin resistance. To determine the functional significance ofSocs3 in vivo, we generated neural cell−specific SOCS3conditional knockout mice using the Cre–loxP system.Compared to their wild-type littermates, Socs3-deficient miceshowed enhanced leptin-induced hypothalamic Stat3 tyrosinephosphorylation as well as pro-opiomelanocortin (POMC)induction, and this resulted in a greater body weight loss andsuppression of food intake. Moreover, the Socs3-deficient micewere resistant to high fat diet−induced weight gain andhyperleptinemia, and insulin-sensitivity was retained. Thesedata indicate that Socs3 is a key regulator of diet-inducedleptin as well as insulin resistance. Our study demonstrates thenegative regulatory role of Socs3 in leptin signaling in vivo,and thus suppression of Socs3 in the brain is a potentialtherapy for leptin-resistance in obesity.

It was initially proposed that leptin has a primary role as an anti-obesity hormone; however, high concentrations of serum leptin levelsare often associated with obesity, suggesting the presence of leptinresistance1−3. The leptin receptor is highly expressed in the hypothala-mus and belongs to the cytokine-receptor superfamily that activatesthe Janus kinase−signal transducers and activators of transcription(Jak-Stat) pathway4. Stat3 is apparently essential for leptin regulationof energy balance5−7. Several mechanisms for leptin resistance havebeen documented, including impaired leptin transport across theblood-brain barrier8 and the presence of negative regulators of leptinsignaling, such as the tyrosine phosphatases SH2-containing proteintyrosine phospatase-2 (SHP-2) (ref. 9) and protein-tyrosine phos-phatase 1B (PTP1B)10−12 as well as the suppressors of cytokine signal-ing family of molecules13. Peripheral leptin administration to micerapidly induces Socs3 mRNA in hypothalamic regions14. Socs3 bindsto the phosphorylated leptin receptor through its Src homology-2

(SH2) domain and inhibits Jak tyrosine kinase activity through its N-terminal kinase inhibitory region, which functions as a pseudo-substrate13,15,16. Furthermore, the C-terminal Socs box recruits theubiquitin-transferase system and may mediate the degradation ofreceptor-Jak complexes17. Thus, Socs3 may be an important regulatorof leptin signaling and could be involved in leptin-resistance. However,because these conclusions were based on the analysis of cell lines, thephysiological significance of Socs3 in leptin signaling and its impacton obesity remained an open question.

Because Socs3-deficient mice die during embryonic development asa result of placental defects18, a conditional knockout approach wasused to determine the tissue-specific roles of Socs3 (ref. 19; Fig. 1a). Togenerate a neural cell−specific deletion, cre transgenic mice, in whichCre recombinase is expressed specifically in neural precursor cellsunder the nestin promoter-enhancer (crenes)20 or synapsin promoter(cresyn)21, were crossed with Socs3flox/flox mice. Genomic DNAextracted from the brain was analyzed with PCR to verify deletion ofSocs3. A 250−base pair band corresponding to the deleted Socs3 allele(Socs3del) appeared in the PCR analysis of brain DNA fromcrenesSocs3flox/flox (NesKO) mice and cresynSocs3flox/flox (SynKO) micebut not in the DNA from control Socs3flox/flox (wild type) mice (Fig. 1b). In these two types of conditional knockout mice, we con-firmed Socs3 deletion in the hypothalamus and cerebrum, but not inother tissues including heart, liver, kidney muscle and fat(Supplementary Fig. 1 online). RT-PCR analysis also indicated thatSocs3 expression was greatly reduced in the brain but not in the othertissues examined (Supplementary Fig. 1 online).

After leptin administration, Socs3 is exclusively expressed in thearcuate nucleus (ARC) of the hypothalamus, which coexpress as leptinreceptors and neuropeptides that are considered to be essential in theregulation of food intake14. Consistent with previous reports, our realtime RT-PCR analysis revealed that leptin treatment led to a twofoldincrease in Socs3 mRNA in wild-type mice (Fig. 1c). However, expres-sion of Socs3 mRNA in NesKO and SynKO mice was much lower thanthat in wild-type mice, irrespective of leptin administration (Fig. 1c).Immunohistological analysis with an antibody to Socs3 confirmed amarked induction of Socs3 protein in the ARC of the brain in

1Division of Molecular and Cellular Immunology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.2Department of Molecular Genetics, Institute of Life Science, Kurume University, 2432-3 Aikawa-machi, Kurume 839-0861, Japan. 3Institute of Molecular Medicineand Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093-0641, USA. 4Cardiovascular Research Institute andThe Third Department of Internal Medicine, Kurume University, 67 Asahi-machi, Kurume 830-0011, Japan. Correspondence should be addressed to A. Y.([email protected]).

Published online 20 June 2004; doi:10.1038/nm1071

Socs3 deficiency in the brain elevates leptin sensitivity andconfers resistance to diet-induced obesityHiroyuki Mori1, Reiko Hanada2, Toshikatsu Hanada1, Daisuke Aki1, Ryuichi Mashima1, Hitomi Nishinakamura1,Takehiro Torisu1, Kenneth R Chien3, Hideo Yasukawa4 & Akihiko Yoshimura1

NATURE MEDICINE VOLUME 10 | NUMBER 7 | JULY 2004 739

©20

04 N

atur

e P

ublis

hing

Gro

up

http

://w

ww

.nat

ure.

com

/nat

urem

edic

ine

Page 2: Socs3 deficiency in the brain elevates leptin sensitivity and confers resistance to diet-induced obesity

L E T T E R S

wild-type mice after the leptin injection, but only a marginal level ofinduction in the two brain-specific Socs3-deficient mice (Fig. 1d).

To clarify the effects of Socs3 deficiency on leptin-induced Stat3 acti-vation, recombinant leptin was injected into mice and the hypothala-mus was examined with immunoblotting and immunofluorescencestaining with phospho-specific (Tyr705) Stat3 antibodies. Stat3 phos-phorylation in the hypothalamus was evident within1 h, after leptininjection but decreased thereafter in wild-type mice (Fig. 2a,b).However, Stat3 activation 1 h after leptin stimulation was significantlygreater in both NesKO and SynKO mice com-pared to their control littermates, and phos-phorylation was sustained for much longer.The ARC of the hypothalamus showed anincrease in both the number and intensity ofneurons positively staining with phospho-

Stat3 (Fig. 2c). Prolonged activation of Stat3in knockout mice was confirmed withimmunofluorescence microscopy (Fig. 2c). Astudy that used mice carrying a mutant leptinreceptor lacking the Stat3 binding siterevealed that Stat3 plays a major role in stim-ulating the catabolic pathways by inducingexpression of the pro-opiomelanocortin(POMC) product α-MSH6,22. The level of

POMC induced by leptin administration was greater in Socs3-deficient mice than in wild-type mice (Fig. 2d). These data indicatethat Socs3 is a negative regulator of leptin-induced Stat3 signaling inthe hypothalamus.

We then examined the effect of Socs3 deletion on body weight lossinduced by leptin. Although leptin at a dose of 1.5 µg/g body weightended to cause a slight decrease in body weight in wild-type mice, thesame dose of leptin infused into either of the two types of neuron-specific Socs3-deficient mice resulted in greater weight loss (Fig. 3a).

740 VOLUME 10 | NUMBER 7 | JULY 2004 NATURE MEDICINE

Tail (primer a, b)

Brain

WT Socs3

Socs3del

Socs3 flox

WT Socs3(primer a, b)

Brain(primer a, c)

Socs3 flox

Socs3flo

x/flox

NesKOSyn

KO

280 bpa b c

Exon 1

380 bp

loxP loxP

a c250 bp

Socs3del

Socs3 flox

WT Socs3

Soc

s3 m

RN

A le

vels

Leptin – + – + – +0

0.5

1

1.5

2

2.5

WT NesKO SynKO

3V

ME

3V

3V

3V

3V

3V

ME

ME

ME ME

ME

WT NesKO SynKO

PBS

Leptin

Socs3flo

x/+

a b

c d

Exon 2

Figure 1 Generation of brain-specific Socs3-deficient mice. (a) Schematic presentation of thewild-type (WT) locus, targeted locus (Socs3flox)and deleted locus (Socs3del). (b) PCR analysis ofgenomic DNA from tails and brain. WT, wild-type.(c) Expression of Socs3 mRNA in thehypothalamus as determined by real-time RT-PCR. The hypothalami of the mice were obtained3 h after leptin (+) or PBS (–) administration.Each group was normalized to the GAPDH values.Mean ± s.e.m. data from two independentmeasurements are shown. WT, wild-type. (d) Immunostaining for Socs3 in thehypothalamus after leptin or PBS injection. 3V, third ventricle; ME, median eminence. WT, wild-type. Scale bar, 100 µm.

3V

ME

3V

3V

3V

3V

3V

ME

ME

ME ME

ME

pStat3

Stat3

pStat3

Stat3

Leptin (h)

Leptin (h)

NesKO WT

pSta

t3 (

%)

400

300

200

100

0

NesKO WT SynKO WT

WT NesKO SynKO

PBS

Leptin

Pom

c m

RN

A l e

vels

NesKO WT

0

2

4

6

10

8

0 1 6 12 0 1 6 12(h)

cont.0 1 6 12 0 1 6 12

(h)0 1 6 12 0 1 6 12 0 1 6 12 0 1 6 12

*** *

*

NesKO WT cont.0 1 6 12 0 1 6 12

a b

c d

Figure 2 Enhanced leptin-induced Stat3activation in the hypothalamus of Socs3-deficient mice. (a) Western blot analysis of Stat3phosphorylation in the hypothalamus of Socs3knockout and wild-type (WT) littermates afterleptin injection (3.0 µg/g body weight). pStat3,phosphorylated Stat3. cont. (control).Representative data from two independentanimals for each time point are shown. (b) Timecourse of phosphorylated Stat3:total Stat3 ratioafter leptin injection (mean ± s.e.m.), asdetermined by densitometry (n = 5 per group).*P < 0.01. WT, wild-type. (c) Immunofluorescentdetection of phosphorylated Stat3 in the ARC 3 hafter leptin injection. 3V, third ventricle; ME,median eminence. WT, wild-type. Scale bar, 100µm. (d) Effects of leptin on Pomc mRNA levels inthe hypothalamus, as determined by real timeRT-PCR. Values normalized to GAPDH areexpressed as a percentage change from thefasted control littermates (n = 5 per group). WT, wild-type. *P < 0.05.

©20

04 N

atur

e P

ublis

hing

Gro

up

http

://w

ww

.nat

ure.

com

/nat

urem

edic

ine

Page 3: Socs3 deficiency in the brain elevates leptin sensitivity and confers resistance to diet-induced obesity

L E T T E R S

Food intake was also suppressed more in NesKO and SynKO mice thanin control wild-type mice (Fig. 3b). Similar effects of Socs3 deletion onbody weight loss and food intake were observed in both male andfemale mice. Pair-feeding experiments indicated that the leptin-induced body weight loss observed in knockout mice was due to areduced food intake (data not shown). Greater body weight loss inSocs3-deficient mice than in wild-type mice was observed throughouta range of leptin doses (0.5−4.5 µg/g body weight, Supplementary Fig.2 online). Circulating leptin levels after leptin injection did not differbetween wild-type and knockout mice (Supplementary Fig. 3 online).These data suggested that Socs3 deletion in the brain resulted in anenhanced leptin signaling, which led to a greater suppression of foodintake and body weight loss.

To examine the role of Socs3 in obesity-associated leptin resistance,mice were placed on a high fat diet (HFD). During 22 weeks on theHFD, both male and female wild-type mice rapidly gained weight (Fig. 4a). However, Socs3-deficient mice on the HFD gained signifi-cantly less weight compared to their wild-type littermates (Fig. 4a,b).Although the snout-anus length did not significantly differ (data notshown), the weight of each fat pad in Socs3-deficient mice on the HFDwas significantly lower than that of wild-type mice (Fig. 4c). Socs3-deficient mice also ingested significantly less food than their wild-typelittermates (Fig. 4d). As expected, after 22 weeks on the HFD, plasmatriglyceride and free-fatty acid levels were significantly lower in condi-tional knockout mice than in wild-type mice (Supplementary Fig. 4online). Plasma leptin levels in wild-type mice on the HFD were

NATURE MEDICINE VOLUME 10 | NUMBER 7 | JULY 2004 741

3V

3V

ME

* NesKO �WT �

NesKOWT

*

Leptin 1.5 µg/g BW/d

∆B

W/B

W x

100

(%)

5

0

–5

–10

–15

��

1 2 3 4 5 6 7Day

Leptin 1.5 µg/g BW/d

∆B

W/B

W x

100

(%)

5

0

–5

–10

–151 2 3 4 5 6 7

Day

�WT �

WT

SynKO

SynKO��

1 2 3 4 5 6 7Day

1 2 3 4 5 6 7Day

2

1

0

–1

–2

–3

Leptin 1.5 µg/g BW/d Leptin 1.5 µg/g BW/d

**

NesKO

WT

SynKO

WT

∆ fo

od in

take

(g)

∆ fo

od in

take

(g)

2

1

0

–1

–2

–3

**

**

a b

Figure 3 Increased effects of leptin on body weight loss and food intake in Socs3-deficient mice. (a) Effect of leptin on body weight. Body-weight change(∆BW) for male NesKO or SynKO mice (filled squares) and wild-type (WT) littermates (open squares) as well as female NesKO or SynKO mice (filled circles)and wild-type littermates (open circles) was measured daily (n = 5 per group). Data are mean ± s.e.m. *P < 0.05. (b) Effect of leptin on food intake. Foodintake was measured for 7 days before leptin administration (for base line) and daily after leptin injection (n = 10 per group). WT, wild-type. Data are mean ± s.e.m. *P < 0.05.

* **

**

350

300

250

200

150

100

50

Glu

cose

(m

g/dl

)

NesKOWT

0 30 60 12090 Time (min)

*** *

NesKOWT

Glu

cose

(%

of i

nitia

l val

ue)

0 30 60 12090 Time (min)

140

120

100

80

60

7 d

food

inta

ke (

g)

� �

27.5

25

22.5

20

17.5

NesKO WTSynKO WT

NesKO WTSynKO WT

* * * * *

NesKO SynKO WT

*

WT

Lept

in (

ng/m

l)8

6

4

2

0

*

*

* **

*

*

*

NesKO WTSynKO WT

NesKO WTSynKO WT

NesKO WTSynKO WT

NesKO WTSynKO WT

Gonadal Retroperitoneal Mesenteric Inguinal

5

4

3

2

1

0

Fat p

ad w

eigh

t (%

of B

W)

*

BW

(g)

40

35

30

25

20

15

n = 15

n = 15

*

NesKO �

WT �

8 10 12 14 16 18 20 22 24 26 28 30

****

NesKO WTSynKO WT

NesKO WTSynKO WT

NesKO WTSynKO WT

NesKO WTSynKO WT

Male Female Male Female

BW

(g)

40

30

20

10

0

Low fat diet High fat diet

Week

10

35

30

25

20

15

BW

(g)

WT �NesKO �

n = 15

n = 15

8 10 12 14 16 18 20 22 24 26 28 30Week

a

b c

d e

f g

Figure 4 Attenuation of diet-induced obesity and insulin-resistance in brain-specific SOCS-deficient mice. (a) Body weight (BW) change in response to aHFD for NesKO mice (filled squares) and wild-type (WT) littermates (open squares). n = 15 per group. *P < 0.05. (b) Average BW of mice fed on a low fatdiet (n = 10) or HFD (n = 13−15) for 22 weeks. WT, wild-type. (c) Weights of different fat deposits (percentage of BW) from three intra-abdominal fat padsand the inguinal fat pad (n = 4, female). *P < 0.05. WT, wild-type. (d) Cumulative food intake over 7 days on a HFD (n = 5). *P < 0.05. WT, wild-type.(e) Leptin levels in plasma taken from the retro-orbital sinuses of male mice fed on a HFD (n = 15); *P < 0.05. (f,g) Glucose (f, n = 8) and insulin (g, n = 11)tolerance test in male mice. Data are mean ± s.e.m. *P < 0.05.

©20

04 N

atur

e P

ublis

hing

Gro

up

http

://w

ww

.nat

ure.

com

/nat

urem

edic

ine

Page 4: Socs3 deficiency in the brain elevates leptin sensitivity and confers resistance to diet-induced obesity

L E T T E R S

∼ 5 ng/ml, suggesting the development of hyperleptinemia and leptinresistance, whereas Socs3-deficient mice showed lower leptin levels,which were similar to the levels seen under normal diet conditions(Fig. 4e). These data show that wild-type mice on the HFD developedobesity and leptin resistance. In contrast, Socs3-deficient mice on theHFD showed a reduced appetite and body weight gain and leptinresistance was rescued in these mice.

Leptin can improve insulin resistance and diabetes in a mousemodel of lipoatrophic diabetes23 and so we examined insulin sensitiv-ity in 28- to 30-week-old NesKO mice and control littermates on anHFD using intraperitoneal glucose- and insulin-tolerance tests. Abolus of glucose was more rapidly cleared by NesKO mice than controlmice (Fig. 4f). Hyperglycemia was more pronounced in the wild-typeanimals than in Socs3-deficient mice at all time points after glucoseadministration. The insulin tolerance test revealed that wild-type miceon the HFD were insulin-resistant, whereas Socs3-deficient mice weresensitive to insulin (Fig. 4g). Taken together, these results demonstratethat ablation of Socs3 expression in neurons ameliorates HFD-induced insulin resistance.

The improvement of glucose tolerance and insulin sensitivity maybe explained by the reduced adiposity in Socs3 knockout mice.However, adiposity is also regulated by brain insulin24, and insulinreceptor gene deletion in the brain resulted in increased food intakeand diet-induced obesity25. Socs3 inhibits insulin signaling by induc-ing the degradation of IRS-1 and IRS-2 (ref. 26) or by direct binding tothe insulin receptor27. Therefore, one can not rule out the possibilitythat Socs3 inhibits insulin signaling in the brain and that this influ-ences propensity to diet-induced obesity and peripheral insulin resist-ance. Further study is necessary to address this point.

Our study suggests that excessive energy intake and body fat deposi-tion in response to a HFD leads to the acquisition of resistance to bothleptin and insulin, whereas loss of Socs3 expression in the brain ame-liorates diet-induced obesity, leptin resistance and even insulin resist-ance. This raises the possibility that the Stat3-Socs3 system in the brainplays a major role in regulating the amount of body fat. This work alsomakes brain Socs3 a potential therapeutic target for the treatment ofleptin resistance, type 2 diabetes and obesity.

METHODSGeneration of conditional Socs3-deficient mice. Socs3flox/flox mice were asdescribed19,28. Nestin-Cre mice expressing Cre recombinase under the controlof the mouse nestin gene promoter (a gift from H. Okano, Keio University,Kanagawa, Japan and S. Noguchi, Meiji Institute of Health Science, Meiji MilkProducts) were as described20,26 . Synapsin1-Cre mice21 were from The JacksonLaboratory. Nestin-Cre mice and Synapsin1-Cre mice were crossed toSocs3flox/flox mice to generate offspring carrying both the cre gene and the heterozygous loxP-flanked Socs3 mutation (Nes-creSocs3flox/+ or Syn-creSocs3flox/+). These mice were further mated with Socs3flox/flox mice, and Nes-creSocs3flox/flox or Syn-creSocs3flox/flox mice were generated. Socs3flox/flox

mice were generated as 129/Bl6 mixed background, and then backcrossed threetimes into C57Bl/6. Nestin-Cre and synapsin-Cre transgenic mice were back-crossed into C57Bl/6 mice at least five times. All animals used in this study weregenerated by crossing creSocs3flox/flox (knockout) and Socs3flox/flox (wild-type)mice and we used littermates for experiments. Genotyping was done withprimers as described19,27 (Fig. 1a). Mice were housed in a 12 h light:dark cycleand temperature-controlled environment. Animal care and all experimentswere conducted in accordance with the institutional guidelines of the KyushuUniversity Graduate School of Medicine.

HFD and leptin administration. Male and female wild-type and Socs3 knock-out mice were fed on a normal diet (low fat: 6% calories from fat). Then 8-week-old NesKO or SynKO and wild-type mice were placed on a HFD com-posed of 23% calories from fat (Oriental Kobo) for up to 30 weeks. For a leptin

administration study, mice from 18 to 26 weeks of age were individually caged.Murine leptin (PeproTech) (0.5−4.5 µg/g body weight) was injected intraperi-toneally twice daily (at 08:30 and at 19:30) for 3 days. Body weight and foodintake were measured daily from 08:00 to 08:30.

Detection of hypothalamic Stat3 and Socs3. Murine leptin (3.0 µg/g bodyweight) or PBS was administered intraperitoneally to mice that had fasted for18 h. After injection, the mice were killed, at 08:30−10:30: the hypothalamuswas isolated and lysed, as described6. Lysates were resolved with SDS-PAGE andmembranes were immunoblotted with anti−phospho-Stat3 (Tyr705) and anti-Stat3 (Cell Signaling). A macrophage cell line (RAW) stimulated with LPS for 3 h in vitro was used as positive control. For quantitative analysis, density of theblots for phosphorylated Stat3 and total Stat3 was measured and expressed asratios of phosphorylated Stat3 to total Stat3. For phospho-Stat3 and Socs3immunofluorescence detection, male wild-type and NesKO mice were injectedintraperitoneally with 3.0 µg/g body weight of leptin and perfused withformaldehyde 3 h after leptin treatment. Hypothalamic sections from micewere also immunostained with anti-Socs3 (IBL) or anti-phospho-Stat3 using areported protocol in rats29.

RNA extraction and RT-PCR. Socs3 knockout and control mice were fasted for18 h to suppress leptin levels and then injected with 3.0 µg/g body weight ofleptin. The hypothalamus was isolated from 08:30 to 10:30 and total RNA waspurified, as described6. We quantified the expression of POMC and Socs3mRNA using RT-PCR as described6,28.

Metabolic measurements. Twenty-eight-week-old male mice fed on a HFD for22 weeks were fasted overnight, and blood was collected from the retro-orbitalsinus at 09:00. Blood glucose was measured with an Antsense-II glucometer(Sankyo). The serum leptin concentration was determined with enzyme-linkedimmunosorbent assay (ELISA) kits (Morinaga).

Glucose and insulin tolerance tests. Male mice were used because they are morehormonally stable. For a glucose tolerance test, 28-week-old NesKO mice or lit-termates fed on a HFD were fasted overnight and glucose was administeredintraperitoneally (1.5 mg/g body weight). For the insulin tolerance test,30-week-old male mice were fed ad libidum, and 0.5 mU/g body weight ofhuman insulin (Novolin R, Novo Nordisk) was injected intraperitoneally. Theglucose levels were measured in blood withdrawn from the tail.

Statistical analysis. Data are expressed as the mean ± s.e.m. Statistical signifi-cance was tested with an unpaired two-tailed Student’s t-test and analysis ofvariance (ANOVA). The differences were considered to be significant ifP < 0.05.

Note: Supplementary information is available on the Nature Medicine website.

ACKNOWLEDGMENTSWe thank Y. Kawabata for technical assistance; N. Arifuku, F. Yamaura and Y. Nishifor manuscript preparation; P. Kievit and J.S. Flier for discussions; and M. Oharafor language assistance. Supported by special grants-in-aid from the Ministry ofEducation, Science, Technology, Sports, and Culture of Japan, the Japan HealthScience Foundation, Mochida Memorial Foundation, and the Uehara MemorialFoundation.

COMPETING INTERESTS STATEMENTThe authors declare that they have no competing financial interests.

Received 11 March; accepted 2 June 2004Published online at http://www.nature.com/naturemedicine/

1. Zhang Y. et al. Positional cloning of the mouse obese gene and its human homo-logue. Nature 372, 425−432 (1994).

2. Considine R.V. et al. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N. Engl. J. Med. 334, 292−295 (1996).

3. Ahima, R.S. & Flier, J.S. Leptin. Annu. Rev. Physiol. 62, 413−437 (2000).4. Friedman, J.M. & Halaas, J.L. Leptin and the regulation of body weight in mam-

mals. Nature 395, 763−770 (1998).5. Ghilardi N. et al. Defective STAT signaling by the leptin receptor in diabetic mice.

Proc. Natl. Acad. Sci. USA 93, 6231−6235 (1996).6. Bates S.H. et al. STAT3 signalling is required for leptin regulation of energy bal-

742 VOLUME 10 | NUMBER 7 | JULY 2004 NATURE MEDICINE

©20

04 N

atur

e P

ublis

hing

Gro

up

http

://w

ww

.nat

ure.

com

/nat

urem

edic

ine

Page 5: Socs3 deficiency in the brain elevates leptin sensitivity and confers resistance to diet-induced obesity

L E T T E R S

ance but not reproduction Nature 421, 856−859 (2003).7. Gao Q. et al. Disruption of neural signal transducer and activator of transcription 3

causes obesity, diabetes, infertility, and thermal dysregulation. Proc. Natl. Acad.Sci. USA 101, 4661−4666 (2004).

8. Schwartz, M.W., Peskind, E., Raskind, M., Boyko, E.J. & Porte, D. Jr.Cerebrospinal fluid leptin levels: relationship to plasma levels and to adiposity inhumans. Nat. Med. 5, 589−593 (1996).

9. Carpenter, L.R. et al. Enhancing leptin response by preventing SH2-containingphosphatase 2 interaction with Ob receptor. Proc. Natl. Acad. Sci. USA. 95,6061−6066 (1998).

10. Elchebly, M. et al. Increased insulin sensitivity and obesity resistance in micelacking the protein tyrosine phosphatase-1B gene. Science 283, 1544−1548(1999).

11. Zabolotny. J.M. et al. PTP1B regulates leptin signal transduction in vivo. Dev. Cell2, 489−495 (2002).

12. Cheng, A. et al. Attenuation of leptin action and regulation of obesity by proteintyrosine phosphatase 1B. Dev. Cell 2, 497−503 (2002).

13. Yasukawa, H., Sasaki, A. & Yoshimura, A. Negative regulation of cytokine signal-ing pathways. Annu. Rev. Immunol. 18, 143−164 (2000).

14. Bjorbaek, C., Elmquist, J.K., Frantz, J.D., Shoelson, S.E. & Flier, J.S.Identification of SOCS-3 as a potential mediator of central leptin resistance. Mol.Cell. 1, 619−625 (1998).

15. Bjorbaek, C. et al. SOCS3 mediates feedback inhibition of the leptin receptor viaTyr985. J. Biol. Chem. 275, 40649−40657 (2000).

16. Eyckerman, S., Broekaert, D., Verhee, A., Vandekerckhove, J. & Tavernier, J.Identification of the Y985 and Y1077 motifs as SOCS3 recruitment sites in themurine leptin receptor. FEBS Lett. 486, 33−37 (2000).

17. Alexander, W.S. Suppressors of cytokine signalling (SOCS) in the immune system.Nat. Rev. Immunol. 2, 410−416 (2002).

18. Roberts, A.W. et al. Placental defects and embryonic lethality in mice lacking sup-

pressor of cytokine signaling 3. Proc. Natl. Acad. Sci. USA 98, 9324−9329(2001).

19. Yasukawa, H. et al. IL-6 induces an anti-inflammatory response in the absence ofSOCS3 in macrophages. Nat. Immunol. 4, 551−556 (2003).

20. Tronche, F. et al. Disruption of the glucocorticoid receptor gene in the nervous sys-tem results in reduced anxiety. Nat. Genet. 23, 99−103 (1999).

21. Zhu, Y. et al. Ablation of NF1 function in neurons induces abnormal developmentof cerebral cortex and reactive gliosis in the brain. Genes Dev..15, 859−876(2001).

22. Thornton, J.E., Cheung, C.C., Clifton, D.K. & Steiner, R.A. Regulation of hypo-thalamic proopiomelanocortin mRNA by leptin in ob/ob mice. Endocrinology 138,5063−5066 (1997).

23. Ebihara, K. et al. Transgenic overexpression of leptin rescues insulin resistanceand diabetes in a mouse model of lipoatrophic diabetes. Diabetes 50, 1440−1448 (2001).

24. Woods, S.C., Lotter, E.C., McKay, L.D. & Porte, D. Jr. Chronic intracerebroventric-ular infusion of insulin reduces food intake and body weight of baboons. Nature282, 503−505 (1979).

25. Bruning, J.C. et al. Role of brain insulin receptor in control of body weight andreproduction. Science 289, 2122−2125 (2000).

26. Rui, L., Yuan, M., Frantz, D., Shoelson, S. & White, M.F. SOCS-1 and SOCS-3block insulin signaling by ubiquitin-mediated degradation of IRS1 and IRS2. J. Biol. Chem. 277, 42394-42398 (2002).

27. Emanuelli, B. et al. SOCS-3 is an insulin-induced negative regulator of insulin sig-naling. J. Biol. Chem. 275, 15985−15991 (2000).

28. Kimura, A. et al. SOCS3 is a physiological negative regulator for granulopoiesis andG-CSF receptor signaling. J. Biol. Chem. 297, 6905−6910 (2004).

29. Munzberg, H., Huo, L., Nillni, E.A., Hollenberg, A.N. & Bjorbaek C. Role of signaltransducer and activator of transcription 3 in regulation of hypothalamic proopi-omelanocortin gene expression by leptin. Endocrinology 144, 2121−2131 (2003).

NATURE MEDICINE VOLUME 10 | NUMBER 7 | JULY 2004 743

©20

04 N

atur

e P

ublis

hing

Gro

up

http

://w

ww

.nat

ure.

com

/nat

urem

edic

ine