10

Click here to load reader

Normalization of obesity-associated insulin resistance through immunotherapy

Embed Size (px)

Citation preview

Page 1: Normalization of obesity-associated insulin resistance through immunotherapy

Normalization of obesity-associated insulin resistancethrough immunotherapyShawn Winer1,5, Yin Chan1,5, Geoffrey Paltser1, Dorothy Truong1, Hubert Tsui1, Jasmine Bahrami2,Ruslan Dorfman4, Yongqian Wang4, Julian Zielenski4, Fabrizio Mastronardi1, Yuko Maezawa1,Daniel J Drucker2, Edgar Engleman3, Daniel Winer3 & H.-Michael Dosch1

Obesity and its associated metabolic syndromes represent a growing global challenge, yet mechanistic understanding of this

pathology and current therapeutics are unsatisfactory. We discovered that CD4+ T lymphocytes, resident in visceral adipose tissue

(VAT), control insulin resistance in mice with diet-induced obesity (DIO). Analyses of human tissue suggest that a similar process

may also occur in humans. DIO VAT-associated T cells show severely biased T cell receptor Va repertoires, suggesting antigen-

specific expansion. CD4+ T lymphocyte control of glucose homeostasis is compromised in DIO progression, when VAT accumulates

pathogenic interferon-c (IFN-c)-secreting T helper type 1 (TH1) cells, overwhelming static numbers of TH2 (CD4+GATA-binding

protein-3 (GATA-3)+) and regulatory forkhead box P3 (Foxp3)+ T cells. CD4+ (but not CD8+) T cell transfer into lymphocyte-free

Rag1-null DIO mice reversed weight gain and insulin resistance, predominantly through TH2 cells. In obese WT and ob/ob (leptin-

deficient) mice, brief treatment with CD3-specific antibody or its F(ab¢)2 fragment, reduces the predominance of TH1 cells over

Foxp3+ cells, reversing insulin resistance for months, despite continuation of a high-fat diet. Our data suggest that the progression

of obesity-associated metabolic abnormalities is under the pathophysiological control of CD4+ T cells. The eventual failure of this

control, with expanding adiposity and pathogenic VAT T cells, can successfully be reversed by immunotherapy.

The incidence of obesity is increasing worldwide, which is quiteproblematic as it is a high-risk factor for development of the metabolicsyndrome—a melange of pathological conditions that include insulinresistance, glucose intolerance and toxicity, hepatic steatosis anddyslipidemia, as well as the risk of developing type 2 diabetes1. Type2 diabetes and obesity both involve genetic and environmentalfactors2, which represent a major global cause of morbidity andmortality, as current nonsurgical therapies are inadequate3.

Together, the above elements contribute to persistent beta cell stress,dysfunction and type 2 diabetes risk. However, progression of meta-bolic syndrome to overt diabetes is not easily predicted; many patientswith metabolic syndrome never convert, whereas some progress onlytransiently. Likewise, while some obese individuals progress to meta-bolic syndrome, others only have mild metabolic abnormalities2,4. Inany case, adipose tissue inflammation is viewed as a promoter ofprogression, but the underlying mechanisms remain unclear, as doesthe nature of what limits or halts progression in individuals withpersisting metabolic syndrome, as well as what drives type 2 diabetesprogression in those who do convert.

Obesity-associated insulin resistance is a core element of type 2diabetes development. The impairment of insulin sensitivity involvesmultiple organs, prominently including hypertrophic adipose tissue

with an associated rise in serum and tissue levels of fatty acids, adipo-kines, and proinflammatory molecules such as interleukin-6 (IL-6)and tumor necrosis factor-a (TNF-a)1,5. TNF-a can be producedby both adipocytes and, in particular, macrophages6,7. Macrophagesaccumulate in both VAT and subcutaneous adipose tissue (SAT).The roles of fat-resident macrophages in obesity-induced insulin-resistance differ, with VAT being the principal driver of metabolicsyndrome progression8–10.

Macrophage activation is modified by T cells. IFN-g–secretingT cells (TH1), as well as those secreting IL-17 (TH17), enhancemacrophage proinflammatory functions by inducing the release ofIL-1, IL-6 and TNF-a. In contrast, anti-inflammatory IL-4 and IL-13–secreting T cells (TH2), as well as CD4+Foxp3+ regulatory T cells,modify macrophage function by inducing their differentiation intoanti-inflammatory, IL-10–secreting M2, or ‘alternatively activated’,macrophages (AAMs)11,12. AAMs are characterized by abundant sur-face expression of macrophage mannose receptor (MMR) and intra-cellular arginase activity13. Increased tissue levels of IL-10 improveinsulin sensitivity in liver and fat14,15, and AAMs in adipose tissuehave been shown to normalize some of the metabolic abnormalitiesassociated with diet-induced obesity in animal models11,15. Strategiesincreasing the number of AAMs or cells that induce them (for

Received 3 March; accepted 9 June; published online 26 July 2009; doi:10.1038/nm.2001

1Neuroscience & Mental Health Program, Research Institute, The Hospital for Sick Children, University of Toronto Departments of Pediatrics & Immunology, Toronto,Ontario, Canada. 2Department of Medicine, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada. 3Departmentof Pathology, Stanford University School of Medicine, Palo Alto, California, USA. 4Program in Genetics and Genomic Biology, Research Institute, The Hospital for SickChildren, University of Toronto, Toronto, Ontario, Canada. 5These authors contributed equally to this work. Correspondence should be addressed to H.-M.D.([email protected]).

NATURE MEDICINE VOLUME 15 [ NUMBER 8 [ AUGUST 2009 921

ART ICL ES©

2009

Nat

ure

Am

eric

a, In

c. A

ll ri

gh

ts r

eser

ved

.

Page 2: Normalization of obesity-associated insulin resistance through immunotherapy

example, TH2 or CD4+Foxp3+ T cells) might be of therapeuticpromise in insulin resistance and type 2 diabetes.

CD4+ and CD8+ T cells enter VAT and SAT of obese mice andhumans16,17. The function of these fat-associated T cells and theirconceivable roles in obesity, glucose homeostasis or both areunknown, nor are their antigen specificities, activation history or Tsublineage profiles known18.

We investigated the impact of T cells on the glucose homeostasis ofC57BL/6 (B6) mice with DIO. We discovered a progressive bias in therecruitment to and expansion of proinflammatory TH1, but not TH17T cells in adipose tissue, with a body mass–dependent, progressivedecline in the proportions of anti-inflammatory CD4+Foxp3+ regu-latory T cells observed in obese mice and humans. We used indepen-dent experimental model systems in mice to delineate several fat-resident immune elements with distinct impacts on weight gain,insulin resistance and glucose homeostasis. Among these, our obser-vation of VAT-specific T cell receptor bias was most unexpected, as itis typically associated with antigen-specific and/or pathogen-specificT cell expansion as well as autoimmunity19–22.

The predominant T cell effect on glucose homeostasis, revealed byCD4+ T cell reconstitution studies in lymphocyte-free DIO Rag1-null

mice, was improvement of glucose tolerance, enhanced insulin-sensi-tivity and lessening of weight gain. We identified the subsets involvedand probed the overall T cell impact in fully developed DIO, usingimmunotherapy with a clinically effective CD3-specific antibody or itsnonmitogenic F(ab¢)2 fragment23. Both therapies substantially nor-malized insulin resistance and glucose homeostasis, selectively restor-ing CD4+Foxp3+ T cell pools in VAT. Treatment was effective formonths after a short course of injections, despite unchanged high-fatdiet consumption. These results suggest a dual role of fat-associatedT lineage cells, which initially mitigate inflammation-induced insulinresistance until VAT-driven TH1 expansion overwhelms this protectiveeffect, a process that can be targeted and corrected therapeutically.

RESULTS

Characterization of fat-associated T cells

We characterized the numbers and subsets of fat-associated T cellsfrom inguinal SAT and epididymal VAT of 14- to 18-week-old high-fatdiet (HFD)-fed and normal chow diet (NCD)-fed B6 mice, consis-tently collecting about 4 � 105 CD3+ T cells per g VAT and SAT. Thefat-associated T cell subset distribution varied somewhat, with a trendtoward higher CD8 to CD4 ratios, in particular in VAT of HFD-fed

Spleen

48.1

52.95.5

16.9Spleen SAT

16.4 11.7 7.9

11.9 29.0

200 3

2

1

0

CD

3+C

D4+

(cel

ls p

er g

× 1

03 )

CD

4+IL

-17+

(cel

ls p

er g

× 1

03 )

CD

4+IF

N-γ

+

(cel

ls p

er g

× 1

03 )

CD

4+F

oxp3

+

(cel

ls p

er g

× 1

03 )

150

100

80

16:1

15:1

10:1

5:1

0

T-be

t/Fox

p3 r

atio

T-be

t/Fox

p3 r

atio

12:1

8:1

4:1

20

15

10

5

0

60

40

20

0

50

SAT

* **

*

*

*

VAT SAT VAT

SAT

BMI (kg m–2)20 25

BMI <25 BMI >30

30 35 40

*

45

VAT SAT VAT

NCDHFD

0

41.1 14.5 35.549.6 37.6

39.3

39.4

35.9 32.4 50.3

CD4 (gated cells)

14.9 41.3 1.45 2.10 1.97

8.1 6.9 16.6

6.3

OVA TCR Vα (CD3+CD4+ gated cells)

3.9 20.7

6.9 4.1 7.8

0.58

6 weeks

Spleen SAT VAT

NCD16 weeks

HFD16 weeks

OT2 spleen(sorted)

Vα: 2 4 6 8 10 12 14 16 18 20

OT2 VAT(sorted)

WT VAT(HFD)

WT VAT(NCD)

WT spleen

2.933.66

22.1

4.9 46.8 7.8 43.848.3 49.5

20.0 29.9

NCDa

c d

f

g

e

b

HFD

NCD

HFD

30

20

NCDHFD

10

Spleen SAT VAT0

CD8

CD4 (gated cells)

CD

4F

oxp3

Fox

p3+ c

ells

(%

of C

D4)

IFN

IL-1

7

CD

3

SAT VAT

VAT

NCD

HFD

Spleen SAT VAT Spleen SAT VAT

Figure 1 Phenotype of fat-associated

T cells. (a) CD4 and CD8 expression onCD3+ cells in spleen, SAT and VAT of B6

HFD-fed and NCD-fed mice. (b)

Proportion of CD4+IFN-g+ (TH1) and

CD4+IL-17+ (TH17) T cells in spleen, SAT

and VAT. (c) Proportion of CD4+Foxp3+

T regulatory cells in spleen, SAT and VAT

(top, FACS plots; bottom, pooled data; *P o 0.03, t test). (d) Total number of CD3+CD4+, CD4+IFN-g+ (TH1), CD4+IL-17+ (TH17) and CD4+Foxp3+

T cells per g of SAT and VAT in NCD-fed and HFD-fed mice (n ¼ 4 or 5 per group, *P o 0.05, t test). (e) Top left, T-bet (TH1) to Foxp3 ratio in human

VAT (r2 ¼ 0.62, P o 0.05). Bottom left, VAT T-bet to Foxp3 ratio in humans with BMI 430 versus BMI o25 (*P o 0.02, t test). Right, Foxp3 (brown)

and T-bet (blue, TH1 cells) in VAT from humans with BMI 430 (top) or BMI o25 (bottom). Scale bar, 50 mm. T-bet to Foxp3 ratios are from 4200 stained

cells from two tissue levels. (f) Flow cytometric analysis of CD3+CD4+ T cells (%) with secondary re-rearranged non-OVA TCRa, as detected by their low

OT2 TCRa to CD3 ratio in spleens, SAT and VAT of 6- or 16-week-old NCD-fed or HFD-fed B6 OT2 mice. (g) Expression of the genes encoding TCR Vaamong CD4+ T cells isolated from spleens of 16-week-old obese WT or OT2 mice and from VAT of HFD-fed and NCD-fed WT and OT2 mice. CD4+ OT2

T cells were identified and sorted as in f. All mice were 14–16-weeks-old, and each contour plot represents three or four independent experiments. All FACS

plots include cell percentages of the corresponding quadrants; histograms represent means ± s.d.

ART ICL ES

922 VOLUME 15 [ NUMBER 8 [ AUGUST 2009 NATURE MEDICINE

©20

09 N

atu

re A

mer

ica,

Inc.

All

rig

hts

res

erve

d.

Page 3: Normalization of obesity-associated insulin resistance through immunotherapy

mice compared to VAT of regular diet controls (Fig. 1a). VAT fromlean and obese mice also contained pools of CD3+CD4�CD8�lym-phocytes (that is, natural killer (NK), NKT and Tg/d lineage cells)24,and these were largely unaffected by obesity (Fig. 1a).

CD4+ effector T cells can generally be subdivided into proinflam-matory cells (TH1, TH17) and anti-inflammatory and regulatorysublineages (TH2, Foxp3+). There were fewer IFN-g–secreting TH1cells in SAT than in VAT from NCD and HFD B6 mice (Fig. 1b). HFDgenerated a progressive IFN-g bias among fat-associated T cells,whereas it diminished the small VAT TH17 sublineage (Fig. 1b).Compared to VAT of NCD-fed mice, the proportion of regulatoryCD4+Foxp3+ T cells in VAT from HFD-fed mice was 70% lower,probably contributing to the proinflammatory VAT profiles of obesemice (Fig. 1c). The ratio of TH1 to Foxp3+ T cells in VAT increasedfrom B1.5:1 in NCD-fed mice to B6.5:1 in DIO mice. In terms ofabsolute numbers, approximately three times more TH1 cells accu-mulated per gram of fat in HFD-fed mice (Fig. 1d), whereas numbersof CD4+Foxp3+ T cells per g of VAT varied little (Fig. 1d). Thenumbers of TH1 cells in adipose tissue thus represent the dynamicvariable affected by diet-induced obesity. The high proportion ofCD4+Foxp3+ T cells in VAT of lean mice, which are progressivelydiluted by IFN-g–producing cells in progressive DIO, suggests onecandidate cellular mechanism by which progressive obesity leads toworsening metabolic control.

Observations in lean and obese humans were strikingly analogous.In VAT from obese humans (Fig. 1e), TH1 cells that express thelineage-specific Tbet transcription factor25 outnumbered Foxp3+

T cells with a ratio of B12:1, whereas the ratio was B6:1 inVAT of lean humans (Fig. 1e). The relative proportion of TH1

to Foxp3+ cells in VAT correlated with bodymass index (BMI) (Fig. 1e, r 2 ¼ 0.62). As inmice, we observed human fat-associatedT cells clustered around blood vessels, somescattered between adipocytes (SupplementaryFig. 1a). We commonly observed Foxp3+ fat-associated T cells in close proximity to mono-

cytes and macrophages (Supplementary Fig. 1b), predicting possibleinteractions with fat-associated monocytes and macrophages.

Tissue-selective lymphocyte accumulation can reflect a number ofmechanisms, including cognate events, involving T cell receptorengagements with its major histocompatibility complex–antigen con-jugates. Although we did not expect to find selective T cell receptor(TCR) engagement of VAT-associated T cells, we decided to formallyrule this possibility out by using ovalbumin (OVA)-specific, OT2 TCRtransgenic mice26. These B6 mice developed metabolic complicationsof DIO (Supplementary Fig. 2a–c). A small proportion of transgenicOT2 T cells can bypass TCRa allelic exclusion and undergo random,secondary Va rearrangements, yielding a second surface TCR that canrecognize non-OVA antigens27. Such cells can be identified throughtheir reduced surface expression of the original, transgenic TCR Varelative to CD3.

There were small pools (4–7%) of T cells with secondary TCRarearrangements in spleens, VAT and SAT of young OT2 mice (Fig. 1f).Beginning at around 6 weeks of age, secondary TCRa rearrangementsslowly and selectively increased thereafter (7–21%) in VAT of all mice,but, in particular, in HFD-fed OT2 mice (Fig. 1f). This VAT tissue-specific effect indicates a strong selective pressure toward a non-OVAantigen(s); that is, an adipose tissue antigen or antigen modificationpresent only in VAT but not in SAT or elsewhere. Thus, unexpectedly,T cell expansion in VAT seems, at least in part, antigen driven.

We sought further support for this notion and PCR amplified eachof the 20 TCR Va–encoding genes in highly purified T cells withsecondary TCR Va rearrangements. These cells had been sorted byFACS from spleens and VAT of obese, 16-week-old OT2 mice; wild-type (WT) B6 spleens served as controls that expressed the expected

a b

c

fg

d e

156 10

*

*

NCD WT

0

0.5

VAT

/SAT

rat

io

1.0

1.5

0

0.5

1.0

1.5

2.0

2.5VATSAT

* *

Fat m

ass

(g)

NCD Rag1-nullHFD WTHFD Rag1-null

NCD WT

0VAT SAT

10

Cel

l dia

met

er (

pixe

ls)

RE

R (

vCO

2/vO

2)F

ood

inta

ke (

g)

Glu

cose

(m

mol

l–1)

20

30

40

50

60

70

80 *

*

NCD Rag1-null NCD WTNCD Rag1-null

NCD WTNCD Rag1-null

HFD WTHFD Rag1-null HFD WT

0

00

0

0

0.5

1.0

Insu

lin (

ng m

l–1)

Glu

cose

(m

mol

l–1)

1.5

2.0

0

5

10

15

20

–10–20–30–40–50–60–70

0 15 30 45 60Time (min)

Glu

cose

cha

nge

(%)

75 90

15 30 45Time (min)

60 75 90

5101520 *

*

**

*

253035

0.51.01.52.02.5

00.20.40.60.8

HFD Rag1-null HFD WTHFD Rag1-null

HFD WTHFD Rag1-null

NCD WT

NCD Rag

1-

null

HFD WT

HFD Rag

1-

null

NCD WT

NCD Rag

1-

null HFD W

T

HFD Rag

1-

null

NCD WT

NCD Rag

1-

null HFD W

T

HFD Rag

1-

null

NCD WT

NCD Rag

1-

null HFD W

T

HFD Rag

1-

null

Age (weeks)14

20

25

30

Wei

ght (

g) 35

40

Figure 2 Impact of lymphocyte deficiency on

weight gain, fat distribution, glucose tolerance

and insulin resistance. (a) Body weights of

Z6-week-old WT and Rag1-null B6 mice on NCD

(n ¼ 20 per group) or HFD (n ¼ 20 per group,

*P o 0.03, Mann-Whitney, WT versus Rag1-null).

(b) Weights of single epididymal VAT and inguinal

SAT pads from 14- to15-week-old WT and Rag1-

null mice on NCD or HFD (left, n ¼ 6 mice per

group, *P o 0.03, t test; right, VAT to SAT ratio,

*P o 0.001, t test). (c) Relative fat cell diameter

(see Online Methods, n ¼ 3 mice per group,

*P o 0.0001, Mann-Whitney). (d) Food intake

(top) and respiratory exchange ratio (RER, bottom)

in HFD-fed WT or Rag1-null mice (n ¼ 4 pergroup). vCO2, carbon dioxide elimination; vO2,

oxygen consumption. (e) Glucose tolerance of

Rag1-null or WT mice on NCD or HFD (n ¼ 10

per group, *P o 0.02, two-way analysis of

variance (ANOVA)). (f) Fasting glucose (left) or

insulin blood concentrations (right) in 14-week-old

HFD-fed and NCD-fed WT or Rag1-null mice

(n ¼ 10 mice per group, *P o 0.05, t test).

(g) Insulin tolerance test (ITT) in 14-week-old WT

Rag1-null mice on NCD or HFD (n ¼ 6–10 per

group, *P o 0.05, two-way ANOVA).

All histograms represent means ± s.d.

ART ICL ES

NATURE MEDICINE VOLUME 15 [ NUMBER 8 [ AUGUST 2009 923

©20

09 N

atu

re A

mer

ica,

Inc.

All

rig

hts

res

erve

d.

Page 4: Normalization of obesity-associated insulin resistance through immunotherapy

full spectrum of TCRa families (Fig. 1g). TCR Va usage in spleen andVAT T cells from obese OT2 mice differed substantially; only some ofthe receptors were systemically shared, including the transgenic Va2

(Fig. 1g). VAT T cells had lost Va3 but gained strong expression of Va5

and Va18 (Fig. 1g). Spectra-typing of the CDR3 region in OT2 TCRa-encoding genes emphasized the strong selection pressures in VAT forvery narrow TCRa diversity and negative pressure against selection ofthe majority of systemic TCR Vas, thus generating an impressive TCRVa bias among VAT T cells, which includes unique Va5 (417 basepairs) and Va18 (393 base pairs) regions (Supplementary Fig. 3).

Motivated by the VAT tissue–specific TCR selection bias and to ruleout possible effects of the TCR transgene, we repeated this analysis butincluded sorted CD4+TCRb+NK1.1� VAT T cells from HFD-fed aswell as NCD-fed B6 WT mice (Fig. 1g). We observed a strong TCR Vabias in VAT of lean (10 of 20 TCR Va families expressed) and, more so,in VAT of HFD-fed mice (only seven of the same ten Va families usedin lean mice) (Fig. 1g). Spectra-typing of the CDR3 region showedmarkedly altered diversity in VAT TCR Va repertoires of DIO versus

lean B6 mice (Supplementary Fig. 4). These results suggest antigen-specific expansion of T cell repertoires in VAT of lean mice, withincreasing bias in progressive obesity, probably driven by adiposetissue changes that occur as a result of obesity. Tissue-driven TCR biasis common in organ-selective autoimmune disorders19–22. However,this process has not previously been associated with obesity or type 2diabetes, and will require identification of the relevant tissue selfepitopes to formally prove autoreactivity.

Metabolic role of T cells in obesity

HFD-fed Rag1-null mice lack lymphocytes, gain more weight andshow greater visceral adiposity than HFD-fed WT mice (Fig. 2a,b).Increased visceral obesity in HFD-fed Rag1-null mice was mostly dueto notable adipocyte hypertrophy (Fig. 2c and Supplementary Fig. 5).There were no significant abnormalities in body weight, adipose tissuemass or adipocyte size in Rag1-null mice on NCD (Fig. 2a–c), norwere there differences in food intake, CO2 output or O2 consumptionbetween HFD-fed Rag1-null and HFD-fed WT mice (Fig. 2d).

d e

f

g

j

Spleen

Rag1-null+ CD8T cells

CD8

CD4

Rag1-null+ CD4T cells

CD

397.1 95.2 95.3

SAT VAT

92.6 94.1 97.1

a

30

20

Glu

cose

(m

mol

l–1)

10

0

20

15

Glu

cose

(m

mol

l–1)

10

5

0

2.0

1.5

Insu

lin (

ng m

l–1)

1.0

0.5

0

2.0

1.5

Insu

lin (

ng m

l–1)

1.0

0.5

0 Wei

ght c

hang

e (g

)

01234520

15

Glu

cose

(m

mol

l–1)

10

5

0

0 15 30 45 60 75 90Time (min)

b

40

30

Wei

ght (

g)

20

10

1.5

1.0

Fat m

ass

(g)

0.5

00

Time after transfer (weeks)2 4

**

Rag1-nullRag1-null + CD8+ T cellsRag1-null + CD4+ T cells

Rag1-

null

Rag1-

null +

CD4+ T

cells

Rag1-

null +

CD8+ T

cells

VATSAT

*

* * * * * *

**

**

c Rag1-nullRag1-null + CD4+ T cellsRag1-null + CD8+ T cells

80

70

60

50

40

Cel

l dia

met

er (

pixe

ls)

30

20

10

0VAT SAT

*

*Rag1-nullRag1-null + CD8+ T cellsRag1-null + CD4+ T cells

Rag1-nullRag1-null + CD8+

Rag1-null + CD4+

h

30

20

Glu

cose

(m

mol

l–1)

10

00 15 30 45 60 75 90

Time (min)

*

Rag1-nullRag1-null + CD4+

Rag1-null + CD4+OT2hi

Rag1-nullRag1-null + CD8+

Rag1-null + CD4+

0

–70

–60

–50

–40

Glu

cose

cha

nge

(%)

–30

–20

–10

0

15 30Time (min)

45 60 75 90

i

*

Rag1-nullRag1-null + CD4+

Rag1-null + CD4+OT2hi

0

–70–60–50–40

Glu

cose

cha

nge

(%)

–30–20–10

0

15 30Time (min)

45 60 75 90

0.6 0.4

VATSpleen

OVA TCR Vα (CD3+CD4+ gated cells)

CD

3

Rag1-

null

+ OT2

hi

+ CD4

+

Rag1-

null

+ OT2

hi

+ CD4

+

Rag1-

null

+ OT2

hi

+ CD4

+

Figure 3 CD4+ T cell grafts reverse obesity-

associated metabolic abnormalities in Rag1-null

mice. (a) FACS plots analyzing purity of

CD3+CD8+ (top) and CD3+CD4+ (bottom) T cells

in spleen, SAT and VAT 2 weeks after transfer

into 12-week-old HFD-fed Rag1-null recipients.All subsequent experiments were performed

2–4 weeks after transfer and compared to non-

transferred, age-matched, HFD-fed Rag1-null

mice. (b) Left, body weights (n ¼ 5 per group) of

recipient mice after transfer (*P o 0.05, t test,

data are from one of four similar experiments).

Right, weights of individual VAT and SAT pads (n ¼ 5 per group); *P o 0.05, t test. (c) Adipocyte diameter after transfer of CD4+ or CD8+ T cells into

HFD-fed Rag1-null mice (n ¼ 3 mice per group, *P o 0.01, Mann-Whitney). (d) Glucose tolerance in HFD-fed Rag1-null recipients after CD4+ T cell

transfer, CD8+ T cell transfer or no transfer (n ¼ 10 per group, *P o 0.0001, two-way ANOVA). (e) Fasting glucose (top) and insulin (bottom) in CD4+ or

CD8+ T cell-transferred HFD-fed Rag1-null mice (n ¼ 5–10 mice per group, *P o 0.03, **P o 0.01, t test). (f) Insulin tolerance in HFD-fed Rag1-null

mice reconstituted with CD4+ T cells, CD8+ T cells or nonreconstituted controls (n ¼ 10, *P o 0.05, two-way ANOVA). (g) FACS plots of OT2 TCR Varearrangements in spleen and VAT, 2 weeks after transfer. The purity of CD4+OT2hi cells transferred was 99.5%. (h,i) Glucose tolerance test (h) and

insulin tolerance test (i) after transfer of CD4+ OT2 TCRahi (CD4+OT2hi) T cells or CD4+ T cells into HFD-fed Rag1-null mice (n ¼ 5 per group,

*P o 0.0001, two-way ANOVA). (j) Fasting glucose (left) and fasting insulin (middle) in CD4+ or CD4+ OT2 TCRahi (OT2hi)-transferred HFD-fed Rag1-null

mice (n ¼ 5 mice per group, *P o 0.05, t test). Right, weight gain after transfer of CD4+ or CD4+ OT2hi TCRahi T cells into HFD-fed Rag1-null mice

(n ¼ 5 mice per group, *P o 0.004, t test). All FACS plots include cell percentages of the corresponding quadrants; histograms represent means ± s.d.

ART ICL ES

924 VOLUME 15 [ NUMBER 8 [ AUGUST 2009 NATURE MEDICINE

©20

09 N

atu

re A

mer

ica,

Inc.

All

rig

hts

res

erve

d.

Page 5: Normalization of obesity-associated insulin resistance through immunotherapy

Glucose and insulin tolerances in 14-week-old, HFD-fed Rag1-nullmice were severely impaired, with fasting glucose at diabetic levels(Fig. 2e,f), fasting hyperinsulinemia (Fig. 2f) and low insulin sensi-tivity upon insulin challenge (Fig. 2g). Rag1-null mice on NCDshowed nonsignificant trends toward poorer glucose tolerance andincreased insulin resistance. However, a significant elevation of fastingblood glucose (Fig. 2e–g) implies that the metabolic role of lympho-cytes may be physiological and occurs even in the absence of ahypercaloric diet.

CD4+ T cells regulate obesity and glucose homeostasis

The DIO Rag1-null phenotype delineated an overall protective role oflymphocytes in weight gain and insulin resistance. To identify lym-phocyte subsets involved in this process, we reconstituted 12-week-oldHFD-fed Rag1-null mice with 5 � 106 CD4+ or CD8+ T cells,

each specifically depleted of CD49b+ NK and NKT cells, CD11b+

monocytes and macrophages and CD19+ and CD45R+ B cellsbefore transfer.

Two weeks after transfer, the cells had populated spleen, SAT andVAT with high purity (B95%, Fig. 3a). CD4+ (but not CD8+) T cellreconstitution lowered weight gain quite notably by 2–4 weeks aftertransfer (Fig. 3b), despite similar food intake, CO2 output and O2

consumption in all mice (Supplementary Fig. 6a,b). Lesser weightgain was apparent in SAT and, even more so, in VAT (Fig. 3b), and itwas associated with smaller adipocyte size in both fat compartments(Fig. 3c and Supplementary Fig. 5), when CD4+ T cell-transferredHFD-fed Rag1-null recipients were compared to CD8+ T cell–trans-ferred HFD-fed Rag1-null recipients or Rag1-null control mice. CD4+

T cell transfer also lowered serum levels of obesity-associated adipo-kines such as leptin, resistin and monocyte chemoattractant protein-1

Rag1-null+ CD4+

+ CD4+STAT6 –

Rag1-null+ CD4+

Spleen

Rag1-null

Rag1-null+ CD8+ T cells

Rag1-null+ CD4+ T cells

Rag1-null+ EGFP– T cells

Fox

p3

CD4 (gated cells)

0

0

10.3 2.5

0.14 0.11

0

0

VAT

6543

Wei

ght c

hang

e (g

)

Rag1-

null

+ CD4

+

+ CD4

+ Foxp

3–

+ CD4

+ IL-1

0–

210

800

600

400

IL-4

(pg

ml–1

)

200

0

HFD WT

Rag1-

null

+ CD4

+

4

3

2

IL-1

3 (n

g m

l–1)

1

0

HFD WT

Rag1-

null

+ CD4

+

15

10

Glu

cose

(m

mol

l–1)

5

0

Rag1-

null

+ CD4

+

+ STAT6–

1.5

1.0

Insu

lin (

ng m

l–1)

0.5

0

4

2

3

∆wei

ght (

g)

1

0

Rag1-

null

+ CD4

+

+ STAT6–

Rag1-

null

+ CD4

+

+ STAT6–

GATA-3 (CD4+ gated cells)

7.8 17.2

+ STAT6 +/++ STAT6 –

2.0

1.5

1.0

Insu

lin (

ng m

l–1)

Rag1-

null

+ CD4

+

+ CD4

+ Foxp

3–

+ CD4

+ IL-1

0–

0.5

0* * *

*

* * * * * *

*

* * **

+ CD4+Foxp3–

+ CD4+IL-10–

00

10

Glu

cose

(m

mol

l–1)

20

30

15 30Time (min)

45 60 75 90

00

10

Glu

cose

(m

mol

l–1)

20

30

15 30Time (min)

45 60 75 90

a b 20

15

10

Glu

cose

(m

mol

l–1)

Rag1-

null

+ CD4

+

+ CD4

+ Foxp

3–

+ CD4

+ IL-1

0–

5

0

* * *

d

e f

g h i

j

c

NCD 2.8 33.7

2.3 15.3HFD

GAT

A-3

Spleen

CD4 (gated cells)

VAT

Spleen0

5

10

15

20

NCD

GAT

A-3

+ c

ells

(%

of C

D4)

25

0

5

10

15

20

CD

4+G

ATA

-3+

(cel

ls g

–1 ×

103 )

25

30

35

VAT NCD HFD

*

HFD

Figure 4 CD4+Foxp3� T cells reverse metabolic abnormalities after transfer. (a) FACS plots of

CD4+Foxp3+ T cells (%) in spleen and VAT three weeks after transfer of purified CD4+ or purified

CD4+Foxp3� (eGFP�, 99.1% pure) cells; one of two similar plots is shown. (b) Change of HFD-fed Rag1-null body weight 3 weeks after transfer of CD4+, CD4+Foxp3� or CD4+IL-10– T cells

(n ¼ 5 per group, *P o 0.01, t test). (c) Glucose tolerance test (n ¼ 5 per group, *P o 0.0001,

two-way ANOVA). (d) Fasting glucose (left, n ¼ 5 mice per group, *P o 0.01, t test) and fasting

insulin (right, n ¼ 5 mice per group, *P o 0.01, t test), 3 weeks after transfer of CD4+,

CD4+Foxp3� or CD4+IL-10– T cells into HFD-fed Rag1-null recipients. (e) Quantification of IL-4

(left) and IL-13 (right) production stimulated by CD3-specific antibody and CD28-specific

antibody treatment of VAT T cells of 16-week-old HFD-fed WT and HFD-fed Rag1-null recipients 3

weeks after transfer of CD4+ T cells (n ¼ 3 per group). (f) FACS plots for CD4+ gated T cells from

VAT of HFD-fed Rag1-null mice that received either CD4+STAT6– (left) or CD4+WT (right) T cells

3 weeks previously. (g) Glucose tolerance 3 weeks after transfer of CD4+ or CD4+STAT6– T cells (n ¼ 5 per group, *P o 0.05, two-way ANOVA).

(h) Fasting glucose (left), fasting insulin (middle) and weight change (right) in HFD-fed Rag1-null recipients 3 weeks after transfer of purified CD4+ or

CD4+STAT6– T cells (n ¼ 5 mice per group, *P o 0.01, t test). (i) Representative FACS plot of CD4+GATA-3+ (%TH2) cells in spleen and VAT of 14- to

16-week-old NCD-fed and HFD-fed B6 mice. (j) Left, pooled data from i (n ¼ 4 per group, *P o 0.03, t test). Right, total number of CD4+GATA-3+ (TH2)

T cells per g of VAT in NCD-fed and HFD-fed B6 mice (n ¼ 4 per group, P 4 0.4, t test). All FACS plots include cell percentages of the corresponding

quadrants; histograms represent means ± s.d.

ART ICL ES

NATURE MEDICINE VOLUME 15 [ NUMBER 8 [ AUGUST 2009 925

©20

09 N

atu

re A

mer

ica,

Inc.

All

rig

hts

res

erve

d.

Page 6: Normalization of obesity-associated insulin resistance through immunotherapy

(MCP-1) (Supplementary Fig. 6c), and it normalized glucose toler-ance, with lower fasting insulin and glucose levels and improvedinsulin sensitivity within 2 weeks (Fig. 3d–f).

To determine the role of cognate, TCR-dependent events, wepurified OVA-specific T cells from OT2 TCR-transgenic B6 mice bysorting cells without secondary rearrangements. Two weeks afteradoptive transfer of these CD4+ OT2 TCR Va

hi lymphocytes intoHFD-fed Rag1-null mice, there were still almost no T cells withsecondary rearrangements in spleens or fat (Fig. 3g). Notably, thepresence of exclusively OVA-specific T cells in VAT failed to improveglucose tolerance (Fig. 3h), insulin sensitivity (Fig. 3i), fasting bloodglucose (Fig. 3j), fasting insulin (Fig. 3j) or body weight (Fig. 3j).These results indicate that CD4+ T cells in VAT mediate the positiveglobal controls of DIO-associated metabolic abnormalities in anantigen-specific fashion.

CD4+Foxp3+ T cells are regulators of inflammation, and CD4+

T cell transfer into HFD-fed Rag1-null mice slowly repopulatedRag1-null VAT with CD4+Foxp3+ T cells, by 2–3% 2 weeks aftertransfer (Fig. 4a). To determine whether CD4+Foxp3+ T cells areessential in the regulation of glucose homeostasis after cell transfer, wesorted and transferred CD4+Foxp3� T cells, using the bicistronic,Foxp3-eGFP–transgenic B6 mouse line, in which Foxp3+ cellsco-express the fluorescent green marker28.

Transferred Foxp3�CD4+ T cells showedlittle (B0.1%) conversion to CD4+Foxp3+

cells in spleen or VAT (Fig. 4a), but recipientmice were as protected from weight gain as

mice receiving full CD4+ T cell grafts from WT hosts (Fig. 4b).CD4+Foxp3� T cells were equally sufficient to improve glucosetolerance, fasting glucose and fasting insulin concentrations(Fig. 4c,d), suggesting that metabolic control after CD4+ T celltransfer does not require CD4+Foxp3+ regulatory T cell function inour adoptive transfer model. Regulatory T cells and inducible type 1regulatory T cells produce IL-10, a major anti-inflammatory cytokine,as part of their effector function29. Adoptive transfer of CD4+ T cellsfrom IL-10–null B6 mice was as protective as full WT CD4+ grafts,indicating that T cell–derived IL-10 is dispensable for the beneficialeffects of CD4+ T cells (Fig. 4b–d).

T cells can acquire a TH2 profile during the extensive homeostaticexpansion that occurs after transfer into Rag1-null mice, and weobserved considerable TH2 cytokine production (IL-4 and IL-13) inVAT-derived T cells of HFD-fed Rag1-null CD4+ T cell recipients(Fig. 4e). Maintenance of TH2 cells is largely dependent on signaltransducer and activator of transcription-6 (STAT6), and STAT6deficiency severely impairs, without completely ablating the TH2sublineage, which can be identified by the GATA-3 transcriptionfactor30–32. To determine the role of TH2 cells in our transfer model,we transferred CD4+STAT6-null T cells into HFD-fed Rag1-null mice.As expected, recipients of CD4+STAT6-null T cells generated reducednumbers of CD4+GATA-3+ (TH2) T cells in VAT compared to mice

IsotypeF

oxp3

Glu

cose

cha

nge

(%)

Wei

ght (

g)

Wei

ght (

g)

Cel

l dia

met

er (

pixe

ls)

Glu

cose

(m

mol

l–1)

Glu

cose

(m

mol

l–1)

Insu

lin (

ng m

l–1)

Insu

lin (

ng m

l–1)

IFN

-γ (

ng m

l–1)

IL-1

3 (p

g m

l–1)

GAT

A-3

IL-1

7 (p

g m

l–1)

Spleen

a

d

f

h i j

g

e

b c20

2.5

0 55 70605040

50

45

40

35

30

25

302010

0

45

35

15

10

5

0

25

1.0

12

9

6

3

0

0.5

0

200

150

100

50

0

200

150

100

50

0

–25

30

20

10

0

–50

–75

2.0

1.5

1.0

0.5

0

15

10

30

20

10

0

Isotype

Isotype

0 15 30 45Time (min)

60 75 90

Anti-CD3

Anti-CD3

*

*

*

*

**

*

** **

Isotype Anti-CD3

Isotype

Time (min)

Time (weeks)

Time (weeks)VAT SAT

Anti-CD3IsotypeAnti-CD3

Isotype Anti-CD3

5

0

CD4 (gated cells)

0 15 30 45 60 75 90

Time (min)0 15 30 45 60 75 90

0 4 8 12 16 20

0 1 2 3 4 5 6 7

8.1 17.7

16.418.3

VAT

Anti-CD3

Glu

cose

(m

mol

l–1)

Glu

cose

(m

mol

l–1)

Isotype

CD4 (gated cells)

5.5

16.7 16.6

18.6

Isotype

F(ab′)2

Isotype F(ab′)2 IsotypeCD4 (gated cells)

F(ab′)2

Isotype

Isotype

VAT

20.7

17.0

F(ab′)2F(ab′)2

Isotype Isotype

Isotype

F(ab′)2

Fox

p3

Spleen

VAT

F(ab′)2 F(ab′)2

F(ab′)2

Figure 5 CD3-specific antibody and its F(ab¢)2fragment improve obesity-induced insulin

resistance. Sixteen-week-old obese (HFD) B6

mice received CD-3–specific antibody (anti-CD3,

a–e), isotype-matched control IgG or CD3-specific

F(ab¢)2 (F(ab¢)2, f–j), maintaining HFD for 6 or 9

weeks. (a) FACS plots (%) of CD4+Foxp3+ T cells

in spleen and VAT 9 weeks after CD3-specific

antibody treatment (one of three similar

experiments is shown). (b) Fasting glucose (top)

and insulin (bottom) after CD3-specific antibody

treatment (n ¼ 8, *P o 0.05, t test). (c,d)

Glucose tolerance (c, n ¼ 8, *P o 0.0004, two-

way ANOVA) and ITT (d, n ¼ 8, *P o 0.0005,

two-way ANOVA) after CD3-specific antibodytreatment. (e) Left, body weights of HFD-fed mice

after CD3-specific antibody treatment (n ¼ 8, *P

o 0.05, t test). Right, adipocyte diameter after

CD3-specific antibody treatment (n ¼ 3 mice per

group). (f) Glucose tolerance (left) (n ¼ 5,

*P o 0.01, two-way ANOVA, one of two similar

experiments), fasting insulin (middle) and fasting

glucose (right) in HFD-fed mice (n ¼ 5, *P o0.04, t test) 6 weeks after F(ab¢)2 treatment.

(g) Body weights of HFD-fed mice after F(ab¢)2treatment. (h) CD4+Foxp3+ T cells in spleen

and VAT of HFD-fed B6 mice 6 weeks after

F(ab¢)2 treatment (one of three similar

experiments). (i) IFN-g, IL-17 and IL-13

concentrations after stimulation of B6 VAT

T cells with CD3-specific antibody plus CD28-

specific antibody 8 weeks after F(ab¢)2 treatment

(n ¼ 3 per group). (j) FACS plot of CD4+ gated,GATA-3–stained VAT T cells 8 weeks after

F(ab¢)2 treatment. All FACS plots include cell

percentages of the corresponding quadrants;

histograms represent means ± s.d.

ART ICL ES

926 VOLUME 15 [ NUMBER 8 [ AUGUST 2009 NATURE MEDICINE

©20

09 N

atu

re A

mer

ica,

Inc.

All

rig

hts

res

erve

d.

Page 7: Normalization of obesity-associated insulin resistance through immunotherapy

receiving WT CD4+ T cells (Fig. 4f). CD4+STAT6-null T cells lackedmost of the positive CD4+ impact on glucose tolerance, fastingglucose, fasting insulin and weight gain, identifying metabolic effectsof TH2 cells in the CD4+ T cell transfer model (Fig. 4g,h).

The implication of TH2 cells, as regulators of glucose homeostasis inthe transfer model, prompted us to compare this subset in VAT ofNCD-fed and HFD-fed WT mice (Fig. 4i,j). The proportions of TH2(CD4+GATA-3+) cells were significantly lower (B50%) in VAT ofHFD-fed mice (Fig. 4i,j); however, the absolute numbers of TH2 cells/gram of VAT remained fairly constant (Fig. 4j). As is the casewith CD4+Foxp3+ T cells, our results suggest that smaller TH2cell proportions in DIO-associated VAT represent a candidatemechanism for worsening metabolic control in progressively obesemice, and strategies that increase VAT TH2 cell content might be oftherapeutic benefit.

Immunotherapy of insulin resistance

The various DIO-associated metabolic parameters relevant here arecomplex, and immune controls probably involve multiple levels andmechanisms. Although the scarcity of CD4+Foxp3+ T cells in DIO-associated VAT implicated these cells in metabolic controls, theadoptive transfer model did not. To gain more insight into thisissue, we used immunotherapy to possibly reset the diet-inducedshifts in VAT T cell constituents, as in vivo CD3-specific antibodyfunction depends on the CD4+Foxp3+ T cell compartment for itslong-term tolerogenic effects23,33.

We injected mitogenic CD3-specific antibody34 (10 mg d�1 over 5 d)into 14-week-old HFD-fed WT mice. This protocol promotes T cellself-tolerance through global, transient (r3 weeks) T cell depletion,paralleled by a selective increase of CD4+Foxp3+ T cell pools in sites oftissue inflammation23,33. Nine weeks after CD3-specific antibodytreatment, HFD mice had near lean levels of CD4+Foxp3+ T cells inVAT (Fig. 5a). Treatment improved fasting glucose and insulin levels

and greatly improved glucose tolerance and insulin sensitivity(Fig. 5b–d). There was a transient weight loss, lasting 3–4 weeksafter CD3-specific antibody injection (Fig. 5e). This was probablyattributable to the cytokine storm after initial T cell activation, withTNF-a a major mediator35,36, as the differences in body weight,adipocyte hypertrophy, food intake, CO2 production and O2 con-sumption disappeared over the following weeks on continued HFD(Fig. 5e and Supplementary Fig. 6d). CD3-specific antibody–mediated normalizing effects on insulin resistance and glucose toler-ance emerged 3–4 weeks after injection and lasted the entire observa-tion period (Z4 months, Supplementary Fig. 7a–d), despitecontinued exposure to the hypercaloric diet. We observed a similarbut slightly less effective impact on metabolic parameters after treat-ment of leptin-deficient ob/ob mice with CD3-specific antibody(Supplementary Fig. 7e–i). Reduced efficiency of CD3-specific anti-body in ob/ob mice correlated with reduced restoration of T regulatorycell numbers in VAT (Supplementary Fig. 7e), probably owing to themultiple immune abnormalities associated with leptin deficiency37.

Adverse effects of the cytokine storm may limit the utility of intactCD3-specific antibody therapy. We therefore tested the non-mitogenicF(ab¢)2 fragment of CD3-specific antibody34. Injection of 150 mg ofF(ab¢)2 per d over 5 d resulted in long-term improvements of glucosetolerance and fasting glucose and insulin concentrations (Fig. 5f)without weight loss (Fig. 5g) in HFD-fed mice. F(ab¢)2 treatment alsorestored CD4+Foxp3+ T cell numbers in VAT (Fig. 5h). Notably, theIFN-g+ TH1 bias and the abundance of other VAT cytokines measuredremained unchanged by F(ab¢)2 therapy (Fig. 5i,j), again pointing to,but not proving, that restoration of VAT T regulatory cells improveslong-term glucose and insulin homeostasis.

CD4+Foxp3+ T cells can induce M2 AAMs12, which secrete IL-10and protect against insulin resistance38. To determine whether therestoration of VAT regulatory T cells by CD3-specific anti-body immunotherapy did, in fact, generate IL-10–secreting M2

HFD

28.6 56.9

28.8

10.5

F4/80 (gated cells)

Isotype F(ab′)2

39.6

27.7

38.7 46.3

36.4

13.5

F4/80 (gated cells)

33.1

24.6

MM

RM

MR

NCD800

2.5 250200150100500

2.01.51.00.5

4

3

2

1

0

0

600

400

IL-1

0 (p

g m

l–1)

IL-1

0 (p

g m

l–1)

MC

P-1

(ng

ml–1

)

TN

F-α

(pg

ml–1

)T

NF

-α (

pg m

l–1)

TN

F-α

(pg

ml–1

)

MC

P-1

(ng

ml–1

)M

CP

-1 (

ng m

l–1)

200

MMR– MMRlo MMRhi MMR– MMRlo MMRhi MMR– MMRlo MMRhi

HFD

NCD

MMRlo MMRhi MMRlo MMRhiMMRlo MMRhi

0

600

400

200

0

800

600

400

300

200

100

0

1.0

0.5

0

2000

200

150

100

50

0IL

-10

(pg

ml–1

)

Isotype F(ab′)2

*

*

Isotype F(ab′)2 Isotype F(ab′)2

a

c d

b

Figure 6 F(ab¢)2 therapy alters VAT-resident macrophage phenotype. (a) MMRhi (%, top gate), MMRlo (middle gate) and MMR� (bottom gate) macrophages

from VAT of 16-week-old HFD-fed or lean NCD-fed B6 mice (representative data from four similar experiments). (b) Quantification of IL-10 (left), MCP-1

(middle) and TNF-a (right) produced by lipopolysaccharide (LPS)-stimulated F4/80+ macrophages sorted from VAT of HFD-fed (top) or NCD-fed (bottom)

mice into MMR�, MMRlo and MMRhi cell populations (one of four similar experiments). We consistently failed to obtain sufficient cell numbers for analysis

of MMR� macrophages from VAT from lean mice. (c) FACS plots from two independent experiments measuring MMR expression in F4/80+ macrophages

from HFD-fed B6 VAT 6 weeks after F(ab¢)2 treatment. (d) Quantification of IL-10 (left), MCP-1 (middle) and TNF-a (right) produced by LPS-stimulated F4/80+ macrophages sorted from VAT 6 weeks after F(ab¢)2 treatment (n ¼ 3 per group, *P o 0.05, t test). All FACS plots include cell percentages of the

corresponding quadrants; histograms represent means ± s.d.

ART ICL ES

NATURE MEDICINE VOLUME 15 [ NUMBER 8 [ AUGUST 2009 927

©20

09 N

atu

re A

mer

ica,

Inc.

All

rig

hts

res

erve

d.

Page 8: Normalization of obesity-associated insulin resistance through immunotherapy

macrophages, we established cytokine profiles of M2 macrophages onthe basis of surface staining of MMR. We distinguished three F4/80+

macrophage populations in VAT of both HFD- and NCD-fed mice:MMR�, MMRlo and MMRhi macrophages (Fig. 6a). Only MMRhi

macrophages expressed IL-10; MMRlo macrophages were the largestsource of MCP-1, and MMR� macrophages expressed MCP-1 andTNF-a, likely representing classical M1 macrophages (Fig. 6b).

Notably, diet had little effect on these cytokine patterns (Fig. 6b).However, HFD-fed mice generated more MMR� macrophages andless IL-10–secreting MMRhi macrophages compared to NCD-fed mice(Fig. 6a). F(ab¢)2 therapy increased the MMRhi pool and reduced theMMR� pool by 6 weeks after treatment (Fig. 6c). This macrophageshift generated an B300% increase in IL-10 production, as measuredin purified VAT macrophages from F(ab¢)2–treated DIO mice(Fig. 6d). F(ab¢)2 therapy seems to have dual functions, increasingboth anti-inflammatory T cell and macrophage compartments in VAT.

DISCUSSION

We identified a fundamental role for CD4+ T lymphocytes in theregulation of body weight, adipocyte hypertrophy, insulin resistanceand glucose tolerance, implicating these cells in the control of diseaseprogression in DIO and the ob/ob form of type 2 diabetes. We revealedthe lymphocyte impact with independent models and experiments,including Rag1-null mice, which develop an overtly diabetic pheno-type on a HFD.

Our evidence maps T cell–mediated metabolic rescue to both,CD4+Foxp3+ and CD4+Foxp3� (for example, TH2) T cell compart-ments. Whereas CD4+Foxp3� cells affected obesity and insulin resis-tance, CD4+Foxp3+ cells primarily regulated insulin resistance,although this was not as apparent in our adoptive transfer models,where TH2 cells emerged as regulators of obesity and insulin resis-tance. Overall, our experiments do not rule out effects of B lympho-cytes or VAT-associated innate lymphocyte subsets (for example, NK,NKT, TCRg/d or CD4�CD8�TCRa/b+ subsets)24. In fact, our pre-liminary data suggest that B cell deficiency in DIO mice is associatedwith improved metabolic control (S.W., Y.C., G.P. and H.-M.D.unpublished data).

We primarily focused on mouse models, but our limited humanstudies support the conclusions we drew from the mouse experiments.VAT of DIO mice and obese humans is characterized by an abnormallyhigh TH1 to Foxp3+ cell ratio; high expression of IFN-g in human VATwas previously associated with increased waist circumference17. IFN-gis a proinflammatory cytokine, and increased numbers of TH1 cells inVAT may contribute to insulin resistance, an observation supported byimproved glucose tolerance in TH1 cell–deficient, IL-12p35–null mice(S.W., Y.C., H.T. and H.-M.D., unpublished data) and in IFN-g–nullmice39. We favor a model where a relatively constant pool ofCD4+Foxp3+ and TH2 fat-associated T cells gradually fails to limitexpanding pools of TH1 cells, leading to a progressively proinflamma-tory environment that promotes insulin resistance. It is alsopossible that T regulatory cell function may be suboptimal in theinflammatory VAT environment, as suggested in autoimmune diseasemodels40. Factors that drive TH1 cell influx, expansion or both in VATare unknown but now appear to include antigen-driven expansion,with a role for classical immune elements such as the TH1chemokine RANTES18.

The precise mechanisms by which CD4+ T cells affect insulinsensitivity require further study. The induction of IL-10–secretingM2 macrophages represents one potential mechanism, although weruled out such a role for T cell-derived IL-10. CD4+Foxp3+ T cellsinduce M2c-type AAMs through production of IL-10 and TGF-b,

whereas TH2 T cells can induce M2a-type AMMs13. AAMs canregulate insulin resistance through IL-10–mediated reversal of TNF-a–induced insulin resistance15. AAMs are present in adipose tissue oflean mice, but, as obesity ensues, there is a shift to proinflammatoryM1 macrophages15. This shift to M1 macrophages is probably a resultof steadily increasing TH1 to Foxp3+ and TH1 to TH2 cell ratios. Infact, we observed increases in VAT M2 macrophages and decreases inM1 macrophages after restoration of CD4+Foxp3+ T cell pools afterF(ab¢)2 immunotherapy.

CD3-specific antibody is a potent, clinically used immunosuppres-sant, and a possible therapeutic in type 1 diabetes34,41. Its immuneeffects are reversible, well understood and easily monitored. The datapresented here suggest that it may be of use in the treatment of insulinresistance. In such individuals, use of CD3-specific antibody may havemultiple benefits, as it is effective in reducing atheroscleroticplaques, common in obesity and insulin resistance syndromes42. Weobserved a similar but slightly less effective course of CD3-specificantibody immunotherapy in diabetic, leptin-deficient ob/ob mice.However, leptin affects multiple pathways in the immune response,including TGF-b signaling, T cell homeostasis and macrophage anddendritic cell maturation43,44. CD3-specific antibody immunotherapyrequires TGF-b secretion by phagocytes, and deficiencies in thispathway may account for somewhat reduced efficiency in leptin-deficient mice45.

Although we focused on the metabolic roles of lymphocytes in VAT,we cannot rule out lymphocyte impact on insulin resistance in otherorgans such as the liver. We also cannot rule out a role for CD8+

T cells in disease development. Although transfer studies show thatCD8+ T cells in isolation have little impact on obesity and associatedinsulin resistance, it is possible that, with CD4+ T cell help, CD8+

T cells and possibly the regulatory CD8+ subsets33 become moreactive participants.

Our perhaps most unexpected discovery was the TCR Va bias inDIO OT2 and B6 mice. Antigen-driven T cell selection and expansionin VAT would be the most likely explanation, substantially supportedby the failure of exclusively OVA-specific T cells to transfer theprotective functions of TH2 and Foxp3+ T cell compartments. Furthervalidation, through identification of candidate adipose tissue epitopes,would imply autoimmunity in obesity, where VAT selective adipocytedeath has, in fact, already been described46.

Collectively, we have shown a previously unknown role for theadaptive immune system in the regulation of obesity, fat distributionand insulin resistance. Our observations identify several T cell popula-tions as physiological regulators in these processes, and they identifyVAT-selective, TCR-dependent, cognate immunity as one elementdriving T cell expansion in VAT. This observation strongly impliesthat VAT incites active T cell immunity. Work is underway to identifythe inciting antigenic epitopes to open a door to globally affordabletolerogenic vaccination strategies against DIO and, possibly, itsassociated metabolic syndromes, including type 2 diabetes.

METHODS

Methods and any associated references are available in the onlineversion of the paper at http://www.nature.com/naturemedicine/.

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

ACKNOWLEDGMENTSThese studies were supported by the Canadian Institutes of Health Research. Y.C.and G.P. were recipients of scholarships from the Canadian Institutes of HealthResearch and the Banting & Best Foundation. Antibodies to OT2 TCR heavy andlight chain were a gift from D. Philpott (University of Toronto).

ART ICL ES

928 VOLUME 15 [ NUMBER 8 [ AUGUST 2009 NATURE MEDICINE

©20

09 N

atu

re A

mer

ica,

Inc.

All

rig

hts

res

erve

d.

Page 9: Normalization of obesity-associated insulin resistance through immunotherapy

AUTHOR CONTRIBUTIONSS.W. and Y.C. conceived of the study and carried out the bulk of the experiments.G.P., D.T., H.T. and Y.M. performed a number of mouse experiments. R.D., Y.W.and J.Z. provided much of the TCR material, F.M. helped with adipocyte analysis,J.B. and D.D. provided metabolic studies, E.E. and D.W. generated the bulk ofhuman data, S.W. and H.-M.D. wrote the manuscript, and H.-M.D. financed andsupervised the project and the peer review process.

COMPETING INTERESTS STATEMENTThe authors declare competing financial interests: details accompany the full-textHTML version of the paper at http://www.nature.com/naturemedicine/.

Published online at http://www.nature.com/naturemedicine/.

Reprints and permissions information is available online at http://npg.nature.com/

reprintsandpermissions/.

1. Kahn, S.E., Hull, R.L. & Utzschneider, K.M. Mechanisms linking obesity to insulinresistance and type 2 diabetes. Nature 444, 840–846 (2006).

2. Despres, J.P. & Lemieux, I. Abdominal obesity and metabolic syndrome. Nature 444,881–887 (2006).

3. Dahabreh, I.J. Meta-analysis of rare events: an update and sensitivity analysis ofcardiovascular events in randomized trials of rosiglitazone. Clin. Trials 5, 116–120(2008).

4. Hanson, R.L., Imperatore, G., Bennett, P.H. & Knowler, W.C. Components of the‘metabolic syndrome’ and incidence of type 2 diabetes. Diabetes 51, 3120–3127(2002).

5. Hotamisligil, G.S. Inflammation and metabolic disorders. Nature 444, 860–867(2006).

6. Kim, J.Y. et al. Obesity-associated improvements in metabolic profile through expan-sion of adipose tissue. J. Clin. Invest. 117, 2621–2637 (2007).

7. Furuhashi, M. et al. Adipocyte/macrophage fatty acid-binding proteins contribute tometabolic deterioration through actions in both macrophages and adipocytes in mice.J. Clin. Invest. 118, 2640–2650 (2008).

8. Kanda, H. et al. MCP-1 contributes to macrophage infiltration into adipose tissue,insulin resistance, and hepatic steatosis in obesity. J. Clin. Invest. 116, 1494–1505(2006).

9. Yuan, M. et al. Reversal of obesity- and diet-induced insulin resistance with salicylatesor targeted disruption of Ikkb. Science 293, 1673–1677 (2001).

10. Weisberg, S.P. et al. CCR2 modulates inflammatory and metabolic effects of high-fatfeeding. J. Clin. Invest. 116, 115–124 (2006).

11. Odegaard, J.I. et al. Macrophage-specific PPARg controls alternative activation andimproves insulin resistance. Nature 447, 1116–1120 (2007).

12. Tiemessen, M.M. et al. CD4+CD25+Foxp3+ regulatory T cells induce alternativeactivation of human monocytes/macrophages. Proc. Natl. Acad. Sci. USA 104,19446–19451 (2007).

13. Martinez, F.O., Sica, A., Mantovani, A. & Locati, M. Macrophage activation andpolarization. Front. Biosci. 13, 453–461 (2008).

14. Cintra, D.E. et al. Interleukin-10 is a protective factor against diet-induced insulinresistance in liver. J. Hepatol. 48, 628–637 (2008).

15. Lumeng, C.N., Bodzin, J.L. & Saltiel, A.R. Obesity induces a phenotypicswitch in adipose tissue macrophage polarization. J. Clin. Invest. 117, 175–184(2007).

16. Rausch, M.E., Weisberg, S., Vardhana, P. & Tortoriello, D.V. Obesity in C57BL/6J miceis characterized by adipose tissue hypoxia and cytotoxic T-cell infiltration. Int. J. Obes.(Lond.) 32, 451–463 (2008).

17. Kintscher, U. et al. T-lymphocyte infiltration in visceral adipose tissue. a primary eventin adipose tissue inflammation and the development of obesity-mediated insulinresistance. Arterioscler. Thromb. Vasc. Biol. 28, 1304–1310 (2008).

18. Wu, H. et al. T-cell accumulation and regulated on activation, normal T cell expressedand secreted upregulation in adipose tissue in obesity. Circulation 115, 1029–1038(2007).

19. Baker, F.J., Lee, M., Chien, Y.H. & Davis, M.M. Restricted islet-cell reactive T cellrepertoire of early pancreatic islet infiltrates in NOD mice. Proc. Natl. Acad. Sci. USA99, 9374–9379 (2002).

20. Turner, S.J., Doherty, P.C., McCluskey, J. & Rossjohn, J. Structural determinants of T-cell receptor bias in immunity. Nat. Rev. Immunol. 6, 883–894 (2006).

21. Oksenberg, J.R. et al. Limited heterogeneity of rearranged T-cell receptor V atranscripts in brains of multiple sclerosis patients. Nature 345, 344–346 (1990).

22. Jenkins, R.N., Nikaein, A., Zimmermann, A., Meek, K. & Lipsky, P.E. Tcell receptor V bgene bias in rheumatoid arthritis. J. Clin. Invest. 92, 2688–2701 (1993).

23. Belghith, M. et al. TGF-b–dependent mechanisms mediate restoration of self-toleranceinduced by antibodies to CD3 in overt autoimmune diabetes. Nat. Med. 9, 1202–1208(2003).

24. Caspar-Bauguil, S. et al. Adipose tissues as an ancestral immune organ: site-specificchange in obesity. FEBS Lett. 579, 3487–3492 (2005).

25. Szabo, S.J. et al. A novel transcription factor, T-bet, directs TH1 lineage commitment.Cell 100, 655–669 (2000).

26. Barnden, M.J., Allison, J., Heath, W.R. & Carbone, F.R. Defective TCR expression intransgenic mice constructed using cDNA-based a- and b-chain genes under the controlof heterologous regulatory elements. Immunol. Cell Biol. 76, 34–40 (1998).

27. Padovan, E. et al. Expression of two T cell receptor a chains: dual receptor T cells.Science 262, 422–424 (1993).

28. Fontenot, J.D., Rasmussen, J.P., Gavin, M.A. & Rudensky, A.Y. A function forinterleukin 2 in Foxp3-expressing regulatory T cells. Nat. Immunol. 6, 1142–1151(2005).

29. Awasthi, A. et al. A dominant function for interleukin 27 in generating interleukin 10–producing anti-inflammatory T cells. Nat. Immunol. 8, 1380–1389 (2007).

30. Jankovic, D. et al. Single cell analysis reveals that IL-4 receptor/Stat6 signaling is notrequired for the in vivo or in vitro development of CD4+ lymphocytes with a TH2cytokine profile. J. Immunol. 164, 3047–3055 (2000).

31. Shimoda, K. et al. Lack of IL-4-induced TH2 response and IgE class switching in micewith disrupted Stat6 gene. Nature 380, 630–633 (1996).

32. Kaplan, M.H., Schindler, U., Smiley, S.T. & Grusby, M.J. Stat6 is required formediating responses to IL-4 and for development of TH2 cells. Immunity 4, 313–319 (1996).

33. Bisikirska, B., Colgan, J., Luban, J., Bluestone, J.A. & Herold, K.C. TCR stimulationwith modified anti-CD3 mAb expands CD8+ T cell population and induces CD8+CD25+

Tregs. J. Clin. Invest. 115, 2904–2913 (2005).34. Chatenoud, L. & Bluestone, J.A. CD3-specific antibodies: a portal to the treatment of

autoimmunity. Nat. Rev. Immunol. 7, 622–632 (2007).35. Alegre, M. et al. Acute toxicity of anti-CD3 monoclonal antibody in mice: a model for

OKT3 first dose reactions. Transplant. Proc. 22, 1920–1921 (1990).36. Alegre, M. et al. Hypothermia and hypoglycemia induced by anti-CD3 monoclonal

antibody in mice: role of tumor necrosis factor. Eur. J. Immunol. 20, 707–710 (1990).37. Claycombe, K., King, L.E. & Fraker, P.J. A role for leptin in sustaining lymphopoiesis

and myelopoiesis. Proc. Natl. Acad. Sci. USA 105, 2017–2021 (2008).38. Zeyda, M. & Stulnig, T.M. Adipose tissue macrophages. Immunol. Lett. 112, 61–67

(2007).39. Rocha, V.Z. et al. Interferon-g, a TH1 cytokine, regulates fat inflammation: a role for

adaptive immunity in obesity. Circ. Res. 103, 467–476 (2008).40. Korn, T. et al. Myelin-specific regulatory T cells accumulate in the CNS but fail to

control autoimmune inflammation. Nat. Med. 13, 423–431 (2007).41. Herold, K.C. et al. Anti-CD3 monoclonal antibody in new-onset type 1 diabetes

mellitus. N. Engl. J. Med. 346, 1692–1698 (2002).42. Steffens, S. & Mach, F. Adiponectin and adaptive immunity: linking the bridge from

obesity to atherogenesis. Circ. Res. 102, 140–142 (2008).43. Macia, L. et al. Impairment of dendritic cell functionality and steady-state number in

obese mice. J. Immunol. 177, 5997–6006 (2006).44. Kumpers, P. et al. Leptin is a coactivator of TGF-b in unilateral ureteral obstructive

kidney disease. Am. J. Physiol. Renal Physiol. 293, F1355–F1362 (2007).45. Perruche, S. et al. CD3-specific antibody–induced immune tolerance involves trans-

forming growth factor-b from phagocytes digesting apoptotic T cells. Nat. Med. 14,528–535 (2008).

46. Strissel, K.J. et al. Adipocyte death, adipose tissue remodeling, and obesity complica-tions. Diabetes 56, 2910–2918 (2007).

ART ICL ES

NATURE MEDICINE VOLUME 15 [ NUMBER 8 [ AUGUST 2009 929

©20

09 N

atu

re A

mer

ica,

Inc.

All

rig

hts

res

erve

d.

Page 10: Normalization of obesity-associated insulin resistance through immunotherapy

ONLINE METHODSMice. We purchased WT C57BL/6 mice, ob/ob, Rag1-null, IL-10-null,

STAT6-null, Foxp3-eGFP and OT2 mice from Jackson Laboratories, and we

maintained them in our vivarium in a pathogen-free, temperature controlled,

12-h light and dark cycle environment. The mice received either NCD or HFD

(Research Diets, 60 kcal% fat for the HFD), from 6 weeks of age on. All studies

used males under approved protocols and in agreement with animal ethics

guidelines of the Animal Care Committee at the Hospital for Sick Children,

Research Institute.

Cell transfer experiments. We isolated splenocytes from 8-week-old B6 NCD-

fed mice as previously described43, and we purified CD4+ and CD8+ T cells

(495%, Easy Sep, StemCell Technologies). We gave 12-week-old HFD-fed

Rag1-null mice 5 � 106 T cells intraperitoneally.

Diet and metabolic studies. We weighed all WT and knockout DIO males

regularly. After 8 weeks on HFD, we measured fasting blood glucose and insulin

concentrations (Crystal Chem ELISA). For glucose tolerance tests, we gave

fasted (16 h) mice 0.75–1 g glucose per kg body weight; for ITT mice, we gave

them 0.75 U per kg body weight human regular insulin (Eli Lilly) (2 U per kg in

ob/ob mice). For VAT to SAT ratios, we pooled and averaged the weights of VAT

and SAT fat pads. We measured fat cell diameter with the straight-line tool in

Image SXM software and quantified at least 200 fat cells each from two

different tissue sections per mouse.

Oxymax and food intake studies. We placed mice in individual metabolic

chambers with free access to water and pre-weighed food, measuring O2

consumption, CO2 output and heat at 15-min intervals over 22 h by indirect

calorimetry (Oxymax System, Columbus Instruments). We normalized all

measurements to body weight. Similar studies comparing DIO WT and DIO

Rag1-null mice were performed at Jackson Laboratories.

Fat-associated T cell isolation. We systemically perfused isoflurane-anesthe-

tized mice with PBS, dissected epididymal VAT and inguinal SAT pads (care-

fully avoiding lymph nodes), mashed the tissue and digested with collagenase

(Sigma, 0.2 mg ml�1 in DMEM for 45 min at 37 1C, with manual shaking every

5 min). We pelleted the digests and filtered them (40-mm filter) to enrich fat-

associated T cells.

Flow cytometry and ELISAs. We stained splenocytes or fat-associated T cells for

30 min with commercial (eBiosciences) antibodies to the following proteins

(dilutions in parentheses): CD3 (1 in 150), CD4 (1 in 200), IL-17 (1 in 150),

IFN-g (1 in 100), Foxp3 (1 in 100), IL-10 (1 in 100), BD Biosciences provided

GATA-3 (prediluted) or CD8 (1 in 300). We labeled macrophages with

phycoerythrin-conjugated antibody to F4/80 (1 in 100) (eBioscience) and sorted

them. We used allophycocyanin-streptavidin (1 in 100) (eBioscience) to label

biotinylated MMR (1 in 50) (R&D Systems). For intracellular staining of T cells,

we incubated the cells with phorbol myristate acetate (PMA) (50 ng ml�1) and

ionomycin (750 ng ml�1) for 14 h in HL-1 medium at 37 1C, adding Golgistop

(BD Bioscience) for the last 11 h before FACS plot analysis with FlowJo software.

We measured serum concentrations of TNF-a, IL-6, adiponectin, leptin and

resistin by ELISA in a blinded fashion (AssayGate). We stimulated isolated

fat-associated T cells (200 � 103) with plate-bound antibody to CD3

(1 mg ml�1) plus antibody to CD28 (0.25 mg ml�1) for 72 h in HL-1 medium

(Lonza). We sorted F4/80+ macrophages (150 � 103) from adipose stromal

vascular cells and stimulated them with LPS (100 ng ml�1, 24 h, 10% FBS in

HL-1 medium). For macrophage subset studies, we stimulated 1 � 105 cells per

macrophage subset ex vivo. We followed manufacturer’s protocols for all

ELISAs: IFN-g, IL-10, IL-4 (BD Bioscience), IL-17, IL-13 (R&D Systems),

MCP-1 and TNF-a (eBioscience).

Antibody to CD3. We injected 10 mg CD3-specific antibody (clone 145-2C11,

BD Biosciences) or purified hamster IgG isotype control (eBioscience) in 100 ml

PBS intraperitoneally for 5 consecutive d. For F(ab¢)2 experiments, we injected

150 mg of F(ab¢)2 (Bio X Cell) or isotype control (Bio X Cell) in 150 ml of PBS

intraperitoneally for 5 consecutive d. All mice survived without obvious

complications.

Histology and immunohistochemistry. We fixed VAT and SAT fat pads for

24 h in 10% buffered formalin. Two researchers, including one certified

pathologist (D.W.), analyzed H&E stained samples in a blinded fashion. We

obtained human VAT from mesenteric tissue of surgically removed colons from

patients with colon cancer who gave informed consent under an approved

protocol set by the Stanford Internal Review Board for Human Subjects. For

immunohistochemistry, we used paraffin-embedded sections subjected to

antigen retrieval in pH 6 (Diva Decloaker, Biocare Medical) with the following

antibody dilutions: 1 in 100 mouse antibody to Foxp3 (Abcam), 1 in 100 rabbit

antibody to Tbet (Santa Cruz) and 1 in 100 rabbit antibody to CD14 (Atlas).

Color was produced with Ferangi blue (Biocare Medical) and diaminobenzi-

dine (Vector labs). We determined Tbet to Foxp3 ratios by counting at least 200

stained cells from two different levels of tissue. There was no tumor infiltration

in the areas examined.

T cell receptor studies. We sorted the following cell subsets by FACS into

RNALater stabilization reagent (Qiagen): B100,000 splenic- and B10,000

VAT-derived, rearranged OT2 T cells (CD4+ transgenic TCR Valo) from pooled

DIO OT2 mice (n ¼ 5; 16 weeks old), B5 � 106 splenic- and B50,000 VAT-

derived WT T cells (CD4+ TCR Vb+ NK1.1neg) from pooled DIO B6 mice (n ¼

10; 16 weeks of age) and WT B6 mice (n ¼ 18; 16 weeks of age). We extracted

RNA with the RNA Mini kit (Qiagen) and generated complementary DNA by

random priming (Qiagen).

We used two sets of nascent degenerate sense variable region primers (Va1–

Va20), with antisense primers in the constant region for nested-amplification of

TCRa cDNAs by standard procedures. We analyzed PCR products from the

second amplification round on precast agarose gels (Invitrogen). For high-

resolution analysis, we ran OT2 second-round products, generated with

5¢-carboxyfluorescein–labeled constant region primers, on an ABI Prism 3100

Genetic Analyzer and analyzed profiles with GeneScan software v3.7 (ABI).

Statistical analyses. We assessed statistical significance between two means by

Mann-Whitney and unpaired t tests, using the Welch correction on sample

sizes smaller than six. Curves were analyzed by two-way ANOVA. Statistical

significance was two-tailed and set at 5%.

doi:10.1038/nm.2001 NATURE MEDICINE

©20

09 N

atu

re A

mer

ica,

Inc.

All

rig

hts

res

erve

d.