t-cell profile in adipose tissue is associated with...

17
2637 A lthough obesity has been clearly linked to insulin resistance and type 2 diabetes mellitus, the underlying mechanisms have not been fully elucidated. Accumulating evidence implicates adipose tissue inflammation as a con- tributor to insulin resistance via increased systemic inflam- mation and direct impairment of insulin-mediated glucose uptake. Obese adipose tissue is associated with increases in serum and tissue proinflammatory molecules, such as interleukin-6 (IL-6) and tumor necrosis factor-α, which can induce insulin resistance in vitro. 1,2 Macrophages accu- mulate in adipose tissue in proportion to body mass index (BMI) and adipose cell size, 3,4 and adopt a proinflammatory phenotype in response to diet-induced obesity. 5 It is thought that changes in hypertrophic adipose cells, such as hypoxia, necrosis, chemokine secretion, and fatty acids, attract and activate macrophages, 1 but the specific stimuli and pathways by which this occurs are unclear. Also unclear is the poten- tial role of other immune cells in adipose tissue, and their relationship to localized and systemic inflammation and insulin resistance. Studies in mice indicate that with diet- induced obesity, proinflammatory T-helper 1 (Th1) and CD8 lymphocytes infiltrate visceral adipose tissue (VAT), where they overcome the anti-inflammatory effects of Th2 and T-regulatory (Treg) cells, promoting classical activation of macrophages and systemic insulin resistance or glucose intolerance. 6–13 T cells have been identified in human adipose tissue, 14,15 but there are scant data on T-cell phenotypes or their relationship to systemic inflammation and insulin resis- tance. To test the hypothesis that numerical dominance of proinflammatory CD8 and Th1 over anti-inflammatory Th2 and T-reg cells would be related to systemic inflammation and insulin resistance in humans, we used flow cytometry © 2014 American Heart Association, Inc. Arterioscler Thromb Vasc Biol is available at http://atvb.ahajournals.org DOI: 10.1161/ATVBAHA.114.304636 ObjectiveThe biological mechanisms linking obesity to insulin resistance have not been fully elucidated. We have shown that insulin resistance or glucose intolerance in diet-induced obese mice is related to a shift in the ratio of pro- and anti- inflammatory T cells in adipose tissue. We sought to test the hypothesis that the balance of T-cell phenotypes would be similarly related to insulin resistance in human obesity. Approach and ResultsHealthy overweight or obese human subjects underwent adipose-tissue biopsies and quantification of insulin-mediated glucose disposal by the modified insulin suppression test. T-cell subsets were quantified by flow cytometry in visceral (VAT) and subcutaneous adipose tissue (SAT). Results showed that CD4 and CD8 T cells infiltrate both depots, with proinflammatory T-helper (Th)-1, Th17, and CD8 T cells, significantly more frequent in VAT as compared with SAT. T-cell profiles in SAT and VAT correlated significantly with one another and with peripheral blood. Th1 frequency in SAT and VAT correlated directly, whereas Th2 frequency in VAT correlated inversely, with plasma high-sensitivity C-reactive protein concentrations. Th2 in both depots and peripheral blood was inversely associated with systemic insulin resistance. Furthermore, Th1 in SAT correlated with plasma interleukin-6. Relative expression of associated cytokines, measured by real-time polymerase chain reaction, reflected flow cytometry results. Most notably, adipose tissue expression of anti-inflammatory interleukin-10 was inversely associated with insulin resistance. ConclusionsCD4 and CD8 T cells populate human adipose tissue and the relative frequency of Th1 and Th2 are highly associated with systemic inflammation and insulin resistance. These findings point to the adaptive immune system as a potential mediator between obesity and insulin resistance or inflammation. Identification of antigenic stimuli in adipose tissue may yield novel targets for treatment of obesity-associated metabolic disease. (Arterioscler Thromb Vasc Biol. 2014;34:2637-2643.) Key Words: adaptive immunity C-reactive protein human adipose tissue-specific secretory factor inflammation insulin resistance interleukin-6 interleukin-10 obesity T helper lymphocytes T lymphocytes visceral fat Received on: May 6, 2014; final version accepted on: September 21, 2014. From the Departments of Medicine (T.L., L.-F.L., C.L.), Pathology (D.W., L.T., O.C., H.Z., M.H.Y.C., E.E.), and Surgery (J.M., H.R.), Stanford University, CA. The online-only Data Supplement is available with this article at http://atvb.ahajournals.org/lookup/suppl/doi:10.1161/ATVBAHA.114.304636/-/DC1. Correspondence to Tracey McLaughlin, MD, Division of Endocrinology, Stanford University School of Medicine, 300 Pasteur Dr, Rm S025, Stanford, CA 94305-5103. E-mail [email protected] T-Cell Profile in Adipose Tissue Is Associated With Insulin Resistance and Systemic Inflammation in Humans Tracey McLaughlin, Li-Fen Liu, Cindy Lamendola, Lei Shen, John Morton, Homero Rivas, Daniel Winer, Lorna Tolentino, Okmi Choi, Hong Zhang, Melissa Hui Yen Chng, Edgar Engleman by guest on May 17, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on May 17, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on May 17, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on May 17, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on May 17, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on May 17, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on May 17, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on May 17, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on May 17, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on May 17, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on May 17, 2018 http://atvb.ahajournals.org/ Downloaded from by guest on May 17, 2018 http://atvb.ahajournals.org/ Downloaded from

Upload: hacong

Post on 15-Mar-2018

214 views

Category:

Documents


2 download

TRANSCRIPT

Page 1: T-Cell Profile in Adipose Tissue Is Associated With ...atvb.ahajournals.org/content/atvbaha/34/12/2637.full.pdfevidence implicates adipose tissue inflammation as a ... tial role of

2637

Although obesity has been clearly linked to insulin resistance and type 2 diabetes mellitus, the underlying

mechanisms have not been fully elucidated. Accumulating evidence implicates adipose tissue inflammation as a con-tributor to insulin resistance via increased systemic inflam-mation and direct impairment of insulin-mediated glucose uptake. Obese adipose tissue is associated with increases in serum and tissue proinflammatory molecules, such as interleukin-6 (IL-6) and tumor necrosis factor-α, which can induce insulin resistance in vitro.1,2 Macrophages accu-mulate in adipose tissue in proportion to body mass index (BMI) and adipose cell size,3,4 and adopt a proinflammatory phenotype in response to diet-induced obesity.5 It is thought that changes in hypertrophic adipose cells, such as hypoxia, necrosis, chemokine secretion, and fatty acids, attract and activate macrophages,1 but the specific stimuli and pathways

by which this occurs are unclear. Also unclear is the poten-tial role of other immune cells in adipose tissue, and their relationship to localized and systemic inflammation and insulin resistance. Studies in mice indicate that with diet-induced obesity, proinflammatory T-helper 1 (Th1) and CD8 lymphocytes infiltrate visceral adipose tissue (VAT), where they overcome the anti-inflammatory effects of Th2 and T-regulatory (Treg) cells, promoting classical activation of macrophages and systemic insulin resistance or glucose intolerance.6–13 T cells have been identified in human adipose tissue,14,15 but there are scant data on T-cell phenotypes or their relationship to systemic inflammation and insulin resis-tance. To test the hypothesis that numerical dominance of proinflammatory CD8 and Th1 over anti-inflammatory Th2 and T-reg cells would be related to systemic inflammation and insulin resistance in humans, we used flow cytometry

© 2014 American Heart Association, Inc.

Arterioscler Thromb Vasc Biol is available at http://atvb.ahajournals.org DOI: 10.1161/ATVBAHA.114.304636

Objective—The biological mechanisms linking obesity to insulin resistance have not been fully elucidated. We have shown that insulin resistance or glucose intolerance in diet-induced obese mice is related to a shift in the ratio of pro- and anti-inflammatory T cells in adipose tissue. We sought to test the hypothesis that the balance of T-cell phenotypes would be similarly related to insulin resistance in human obesity.

Approach and Results—Healthy overweight or obese human subjects underwent adipose-tissue biopsies and quantification of insulin-mediated glucose disposal by the modified insulin suppression test. T-cell subsets were quantified by flow cytometry in visceral (VAT) and subcutaneous adipose tissue (SAT). Results showed that CD4 and CD8 T cells infiltrate both depots, with proinflammatory T-helper (Th)-1, Th17, and CD8 T cells, significantly more frequent in VAT as compared with SAT. T-cell profiles in SAT and VAT correlated significantly with one another and with peripheral blood. Th1 frequency in SAT and VAT correlated directly, whereas Th2 frequency in VAT correlated inversely, with plasma high-sensitivity C-reactive protein concentrations. Th2 in both depots and peripheral blood was inversely associated with systemic insulin resistance. Furthermore, Th1 in SAT correlated with plasma interleukin-6. Relative expression of associated cytokines, measured by real-time polymerase chain reaction, reflected flow cytometry results. Most notably, adipose tissue expression of anti-inflammatory interleukin-10 was inversely associated with insulin resistance.

Conclusions—CD4 and CD8 T cells populate human adipose tissue and the relative frequency of Th1 and Th2 are highly associated with systemic inflammation and insulin resistance. These findings point to the adaptive immune system as a potential mediator between obesity and insulin resistance or inflammation. Identification of antigenic stimuli in adipose tissue may yield novel targets for treatment of obesity-associated metabolic disease. (Arterioscler Thromb Vasc Biol. 2014;34:2637-2643.)

Key Words: adaptive immunity ◼ C-reactive protein ◼ human adipose tissue-specific secretory factor ◼ inflammation ◼ insulin resistance ◼ interleukin-6 ◼ interleukin-10 ◼ obesity ◼ T helper lymphocytes

◼ T lymphocytes ◼ visceral fat

Received on: May 6, 2014; final version accepted on: September 21, 2014.From the Departments of Medicine (T.L., L.-F.L., C.L.), Pathology (D.W., L.T., O.C., H.Z., M.H.Y.C., E.E.), and Surgery (J.M., H.R.), Stanford

University, CA.The online-only Data Supplement is available with this article at http://atvb.ahajournals.org/lookup/suppl/doi:10.1161/ATVBAHA.114.304636/-/DC1.Correspondence to Tracey McLaughlin, MD, Division of Endocrinology, Stanford University School of Medicine, 300 Pasteur Dr, Rm S025, Stanford,

CA 94305-5103. E-mail [email protected]

T-Cell Profile in Adipose Tissue Is Associated With Insulin Resistance and Systemic Inflammation in Humans

Tracey McLaughlin, Li-Fen Liu, Cindy Lamendola, Lei Shen, John Morton, Homero Rivas, Daniel Winer, Lorna Tolentino, Okmi Choi, Hong Zhang, Melissa Hui Yen Chng, Edgar Engleman

by guest on May 17, 2018

http://atvb.ahajournals.org/D

ownloaded from

by guest on M

ay 17, 2018http://atvb.ahajournals.org/

Dow

nloaded from

by guest on May 17, 2018

http://atvb.ahajournals.org/D

ownloaded from

by guest on M

ay 17, 2018http://atvb.ahajournals.org/

Dow

nloaded from

by guest on May 17, 2018

http://atvb.ahajournals.org/D

ownloaded from

by guest on M

ay 17, 2018http://atvb.ahajournals.org/

Dow

nloaded from

by guest on May 17, 2018

http://atvb.ahajournals.org/D

ownloaded from

by guest on M

ay 17, 2018http://atvb.ahajournals.org/

Dow

nloaded from

by guest on May 17, 2018

http://atvb.ahajournals.org/D

ownloaded from

by guest on M

ay 17, 2018http://atvb.ahajournals.org/

Dow

nloaded from

by guest on May 17, 2018

http://atvb.ahajournals.org/D

ownloaded from

by guest on M

ay 17, 2018http://atvb.ahajournals.org/

Dow

nloaded from

Page 2: T-Cell Profile in Adipose Tissue Is Associated With ...atvb.ahajournals.org/content/atvbaha/34/12/2637.full.pdfevidence implicates adipose tissue inflammation as a ... tial role of

2638 Arterioscler Thromb Vasc Biol December 2014

to quantify the relative frequency of these T-cell subsets in subcutaneous adipose tissue (SAT) and VAT of healthy overweight or obese subjects who underwent quantitative insulin-sensitivity testing.

Materials and MethodsMaterials and Methods are available in the online-only Data Supplement.

ResultsForty-seven prescreened, nondiabetic, healthy subjects were studied: 20 Roux-en-Y gastric bypass patients with sampling of both SAT and VAT for flow cytometric analysis of T-cell subsets (n=10) and real-time polymerase chain reaction of T-cell–associated cytokines (n=10), and 27 nonsurgical over-weight/moderately-obese subjects with sampling of SAT alone for similar analyses (Table I in the online-only Data Supplement). Thirteen nonsurgical subjects underwent col-lagenase digestion of fresh adipose tissue for flow cytometric analysis of T-cell subsets: 6 of these subjects and an additional 14 frozen tissue specimens from subjects meeting study cri-teria and harvested in an identical manner were included in the real-time polymerase chain reaction analyses of T-cell–associated cytokine expression. Same day peripheral blood for flow cytometry was obtained from all subjects who underwent flow cytometric analysis of adipose tissue. Using the stain-ing and gating strategies depicted in Figure I in the online-only Data Supplement, we determined relative frequencies of T-cell subsets in SAT, VAT, and peripheral blood. Primary analyses were of 2 types: (1) comparison of T-cell subsets and relevant cytokines in SAT versus VAT depots from gas-tric bypass patients (Tables 1 and 2), including relationships between fat depots and peripheral blood T cells (Figure 1) and plasma high-sensitivity C-reactive protein (hsCRP), a marker of systemic inflammation (Figure 2); (2) measurement of the association between T-cell subsets or cytokines and systemic insulin resistance, which was quantified in all nonsurgical subjects and approximately half of the bariatric surgery sub-jects, who were willing to undergo time-intensive metabolic testing before their surgery (Figure 3).

Differences in T-Cell Frequency According to Regional Fat DepotProinflammatory Th1, Th17, and interferon (IFN)-γ+ CD8 cells, expressed as percentage of total CD4 or CD8 T cells,

were markedly increased in VAT relative to SAT, and in both adipose compartments relative to peripheral blood mono-nuclear cells (PBMC; Table 1). This finding can also be seen in Figure 4, depicting intense staining for both CD4 and CD8 in the VAT of an insulin-resistant subject as com-pared with SAT. Interestingly, CD4 and CD8 density in a comparable VAT sample of an IS control with similar BMI was much less dense. Although there was no significant difference in anti-inflammatory Treg% between SAT, VAT, and PBMC, the relative frequency of anti-inflammatory Th2 cells was significantly higher in SAT, but not VAT, as compared with PBMC. T-cell profiles in SAT and VAT were highly correlated with each other with respect to Th1, Th17, CD4, and CD8 cell frequency (Figure 1A–1D), but not Th2 (r=0.46; P=0.19) or Treg (r=−0.13; P=0.76). Of note, the frequency of Th1 in both adipose tissue depots was 10- to 20-fold greater than that of Th2, Treg, and Th17. Given the robust correlations between T-cell subsets in SAT and VAT, we considered the possibility that T-cell profiles might be reflected in peripheral blood. In support of this idea, strong

Nonstandard Abbreviations and Acronyms

BMI body mass index

hsCRP high sensitivity C-reactive protein

IL interleukin

PBMC peripheral blood mononuclear cells

SAT subcutaneous adipose tissue

SSPG steady-state plasma glucose

Th T-helper

Treg T-regulatory

VAT visceral adipose tissue

Table 1. Flow Cytometric Analysis of T-Cell Subsets in Peripheral Blood Mononuclear Cells, Subcutaneous, and Visceral Adipose Tissues in Bariatric Surgery Subjects

T-Cell Subset, %

PBMC (n=10)

SAT (n=10)

VAT (n=10)

P Value* P Value*†‡§

SAT vs VAT

PBMC vs SAT and

VAT

CD4 T cells (total T)

61.9±15 59.6±14.1 53.6±12.2 0.008 §

Th1 (CD4 T) 18.4±13 47±19 63.0±5 0.008 §

Th2 (CD4 T) 0.63±0.6 1.9±1.1 1.4±0.8 0.15 †

Th17 (CD4 T) 0.53±0.5 3.5±1.9 5.6±3.2 0.013 §

Treg (CD4 T) 1.4±1.0 2.2±2.7 3.8±4.3 0.42 NS

CD8 T cells (total T)

32.9±13.8 30.7±9.6 38.4±6.8 0.87 NS

IFNγ+ CD8 cells (CD8 T)

30.9±19.3 69.2±15.6 81.3±7.0 0.04 §

IFN indicates interferon; PBMC, peripheral blood mononuclear cells; NS, nonsignificant value; SAT, subcutaneous adipose tissue; Th, T-helper cells; Treg, T-regulatory cells; and VAT, visceral adipose tissue.

*Paired Student t test.†P<0.05 for PBMC vs SAT; ‡P<0.05 for PBMC vs VAT; and §P<0.05 for

PBMC vs both SAT and VAT.

Table 2. Relative Expression of T-Cell–Associated Cytokines Quantified by Real-Time Polymerase Chain Reaction in Subcutaneous and Visceral Adipose Tissues From Bariatric Surgery Subjects

Cytokine SAT (n=10) VAT (n=10) P Value* (SAT vs VAT)

IL-6 1.3±1.2 2.7±1.6 0.02

IL-10 1.3±0.9 0.32±0.32 0.08

IL-17 1.09±0.54 3.63±3.62 0.04

IFN-γ 1.65±1.9 2.2±1.6 0.57

IFN indicates interferon; IL, interleukin; SAT, subcutaneous adipose tissue; and VAT, visceral adipose tissue.

*Paired Student t test.

by guest on May 17, 2018

http://atvb.ahajournals.org/D

ownloaded from

Page 3: T-Cell Profile in Adipose Tissue Is Associated With ...atvb.ahajournals.org/content/atvbaha/34/12/2637.full.pdfevidence implicates adipose tissue inflammation as a ... tial role of

McLaughlin et al T Cells in Human Adipose Tissue and IR 2639

correlations were seen between PBMC and SAT for Th1, CD4, and CD8 T-cell frequency and, despite the small num-ber, between PBMC and VAT for CD4 (Figure 1E–1H). The correlation between CD8 frequency in VAT and PBMC was of borderline statistical significance (r=0.82; P=0.09;

not shown). Neither Th2 and Treg nor Th17 in PBMC was correlated with frequency in SAT or VAT, confirming that observed correlations were not an artifact from blood con-tamination of tissue. To assess expression of Th cytokine genes in adipose tissue, we performed real-time polymerase

Figure 1. Body mass index–adjusted correlations between T-helper (Th) subsets are expressed as percentage of CD4 cells in subcutane-ous adipose tissue (SAT) and visceral adipose tissue (VAT; A–D), and between Th subsets in adipose tissue and peripheral blood mono-nuclear cells (PBMC; E–H). IFN indicates interferon. Note: Because of limited cell quantity, not all surface markers could be measured on every sample.

Figure 2. Body mass index–adjusted correlations between log plasma high- sensitivity C-reactive protein (hsCRP, mg/dL) and T-helper (Th) subsets are expressed as percentage of CD4 cells in subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT). Note: Because of limited cell quantity, not all surface markers could be measured on every sample.

by guest on May 17, 2018

http://atvb.ahajournals.org/D

ownloaded from

Page 4: T-Cell Profile in Adipose Tissue Is Associated With ...atvb.ahajournals.org/content/atvbaha/34/12/2637.full.pdfevidence implicates adipose tissue inflammation as a ... tial role of

2640 Arterioscler Thromb Vasc Biol December 2014

chain reaction on VAT and SAT from 10 healthy bariatric subjects (Table 2). Expression of IL-6, a marker of Th1 and M1 macrophage activation, was 2-fold greater in VAT than SAT (P=0.02), and IL-17, a marker of Th17 activation (also proinflammatory), was 3-fold greater in VAT than SAT (P=0.04). IL-10, a marker of Th2 and M2 activation and potential mediator of insulin sensitivity, was 4-fold lower in VAT versus SAT (P=0.08).

T-Cell Phenotype: Relationship to Circulating Inflammatory MarkersTo ascertain whether Th profiles in adipose tissue were asso-ciated with systemic inflammation, correlations between Th1 and Th2 in SAT and VAT were performed with respect to both plasma hsCRP and IL-6 concentrations in our cohort. Th1 in VAT was robustly associated with plasma hsCRP (r=0.91; P=0.002), whereas Th2 in VAT was inversely

Figure 4. Representative histology of CD4 and CD8 T cells in human subcutaneous (SAT) and visceral (VAT) adipose tissues in insulin-sensitive (IS) and insulin-resistant (IR) subjects. Brown indicates CD4-positive cells, whereas blue indicates CD8-positive cells. Images are shown at a ×200 magnification. VAT from the IR subject has more CD4 and CD8 cells compared with SAT (left) and VAT from the IS subject (top right). The 60-year old, white female IS subject has body mass index (BMI) 41 kg/m2, steady-state plasma glucose (SSPG) 129 mg/dL, plasma interleukin-6 (IL6) 8.8 pg/mL; and the 56-year old, white female IR subject has BMI 47.3 kg/m2, SSPG 346 mg/dL, and plasma IL6 28.3 pg/mL.

Figure 3. Body mass index–adjusted cor-relations between T-helper (Th2) subsets are expressed as percentage of CD4 cells in subcutaneous adipose tissue (SAT; A) and visceral adipose tissue (VAT; B) and insulin resistance (steady-state plasma glucose [SSPG]), and between SAT interleukin-10 (IL-10) expression and SSPG (C). PBMC indicates peripheral blood mononuclear cells. Note: Because of limited cell quantity, not all surface markers could be measured on every sample.

by guest on May 17, 2018

http://atvb.ahajournals.org/D

ownloaded from

Page 5: T-Cell Profile in Adipose Tissue Is Associated With ...atvb.ahajournals.org/content/atvbaha/34/12/2637.full.pdfevidence implicates adipose tissue inflammation as a ... tial role of

McLaughlin et al T Cells in Human Adipose Tissue and IR 2641

associated with plasma hsCRP (r= −0.75; P=0.03; Figure 2). Th1, but not Th2, in SAT was also strongly associated with plasma hsCRP (r=0.64; P=0.035). Frequencies of the other CD4 and CD8 cells were not associated with hsCRP. Results were unchanged after adjustment for sex. These results point to VAT as having a particularly strong association with sys-temic inflammation, with bidirectional relationships between Th1 and Th2 and plasma hsCRP. Th1, but not Th2, frequency in SAT also correlated with IL-6 concentrations in peripheral blood (Figure II in the online-only Data Supplement; r=0.65; P=0.03; n=15). A similar trend was also demonstrated for Th1 in VAT, although sample size was too limited to draw firm conclusions (r=0.86; P=0.38; n=4).

T-Cell Subsets: Relationship to Insulin ResistanceTo ascertain the relationship between Th profiles and sys-temic insulin resistance, we quantified insulin-mediated glu-cose uptake using the modified insulin suppression test (see Methods in the online-only Data Supplement), denoted by the steady-state plasma glucose (SSPG). Although studies in obese mice have shown that proinflammatory Th1 and CD8 cells play important roles in the adipose tissue inflammation that underlies insulin resistance,6,8 we found no correlation between insulin resistance and the percentages of Th1, CD8, or Th17 cells in SAT, VAT or PBMC. Also in contrast to mice studies,13 anti-inflammatory Treg frequency in SAT and VAT was not significantly associated with SSPG (data not shown). On the contrary, despite lower overall frequency in both SAT and VAT than proinflammatory Th1, the percentage of anti-inflammatory Th2 was inversely correlated with insulin resistance (Figure 3A and 3B). Th2 in PBMC was also sig-nificantly inversely associated with SSPG (Figure 3C). Thus, the results indicate that Th2 bias is independently inversely associated with insulin resistance.

To further explore the relationship between Th2 and insu-lin resistance, we measured the relative expression of IL-10, a Th2-secreted cytokine that potently inhibits proinflamma-tory cytokines and has also been shown to enhance whole body insulin sensitivity in mice.11 Relative expression of this cytokine in the SAT of 20 moderately obese, BMI-matched subjects, was significantly inversely associated with SSPG (P=0.007; Figure 3D), supporting the importance of an anti-inflammatory bias in adipose tissue with regard to protection from systemic insulin resistance. IL-10 is also secreted by cells other than Th2, and the stimulus to and source of IL-10 in adipose tissue are worthy of further investigation.

DiscussionThis study shows, for the first time, that there exists an abun-dant infiltrate of Th1 and Th2 CD4 cells, as well as IFN-γ+ CD8 T cells, in SAT and VAT of healthy overweight and obese individuals, and confirms prior studies demonstrating Th17 cells are present in human fat. Importantly, results demonstrate that subpopulations of T cells differ according to regional fat depot and are associated with both systemic inflammation and insulin resistance. In our healthy overweight/moderately-obese humans, proinflammatory Th1, Th17, and IFN-γ+ CD8 cells were markedly increased in VAT relative to SAT and peripheral

blood. Despite these differences, proinflammatory T-cell fre-quencies in VAT correlated highly with SAT, and with periph-eral blood, indicating that triggers to the adaptive immune response occur in multiple adipose tissue depots, and may even be systemic. Th1 and Th2 cells in VAT bore robust bidirectional relationships to plasma hsCRP, a systemic inflammatory marker that indicates increased risk for both cardiovascular disease and type 2 diabetes mellitus. Furthermore, Th1 frequency in SAT was associated with both plasma hsCRP and IL-6 concentra-tions. Finally, and perhaps most importantly, Th2 frequency in adipose tissue and peripheral blood correlated inversely with insulin resistance, indicating a protective effect. Adipose tissue expression of IL-10, a Th2-related cytokine, was also highly inversely associated with insulin resistance. These relation-ships were independent of obesity per se, and thus the relative dominance of Th1 versus Th2 responses in adipose tissue may account in part for the metabolic heterogeneity that is now well described in overweight and obese humans.18

The present findings represent an important extension to currently published data on the potential role of inflamma-tory cells as mediators of insulin resistance. Although the majority of studies investigating inflammation and insulin resistance have focused on macrophages, 3 recent studies in mice6,8,13 demonstrated an important role for CD4 and CD8 lymphocytes in the development of insulin resistance. We previously demonstrated that CD4 T cells in adipose tissue promote insulin resistance in diet-induced obese mice via dramatic increases in proinflammatory Th1 over static num-bers of anti-inflammatory Treg and Th2. CD4+ T cell transfer, predominantly through Th2 cells, reversed insulin resistance, supporting a causal role. In addition, treatment of obese WT and ob/ob (leptin-deficient) mice with CD3-specific antibody or its F(abʹ)2 fragment, reduced the predominance of Th1 cells over Treg cells, reversing insulin resistance for months.6 Two other studies in mice have shown that Treg and CD8 T cells play opposing roles in adipose tissue inflammation and insulin resistance,8,13 and another showed that that obesity was asso-ciated with selective expansion of the proinflammatory Th17 sublineage.14 Importantly, 2 of these studies6,13 demonstrated little heterogeneity of the T-cell receptor repertoire in VAT, implying localized antigenic stimulation and expansion of antigen-specific T cells. This is plausible because it is known that hypertrophic or hypoxic adipose cells become necrotic, releasing cellular contents and secreted molecules into the extracellular space which serve as antigens to T cells.2,4

Despite the impressive findings in rodent studies, human data on the role of T cells in adipose tissue are scant and there are no studies addressing their potential relationship to insu-lin resistance. To our knowledge, there are only 3 published studies demonstrating CD4 T cell subsets in human adipose tissue using flow cytometry. The first, which examined SAT only, demonstrated increased prevalence of Th17 and Th22 cells in metabolically abnormal versus metabolically normal obese.15 In this study, CD4 T cells staining for IL13 (Th2) and IFN-γ (Th1) were not associated with metabolic profile. It is important to note that the BMI of the metabolically abnormal versus metabolically normal obese subjects in this study was 43.8 versus 34.9 kg/m2, which does not rule out the possibility

by guest on May 17, 2018

http://atvb.ahajournals.org/D

ownloaded from

Page 6: T-Cell Profile in Adipose Tissue Is Associated With ...atvb.ahajournals.org/content/atvbaha/34/12/2637.full.pdfevidence implicates adipose tissue inflammation as a ... tial role of

2642 Arterioscler Thromb Vasc Biol December 2014

that obesity per se contributed to the findings. Furthermore, T cells were expanded in culture for 2 weeks before flow cyto-metric analysis, which leaves open the possibility that T-cell phenotypes were altered by the culture process. The second study,17 which compared SAT and VAT of morbidly obese dia-betic and nondiabetic patients with SAT of nonobese controls, also showed increased frequency of IL17 and IL22-secreting T cells in obese subjects with or without diabetes mellitus as compared with the nonobese controls. Furthermore, exposure of CD4 T cells to macrophage-derived or recombinant IL1B increased production of IL17 and IL22, suggesting cross-talk between immune cells in adipose tissue. The third study demonstrated increased Th17 in VAT of overweight or obese as compared with lean women.18 As in the first study, these results cannot exclude the possibility that alterations in T-cell phenotypes were caused by obesity per se because the com-parisons were made between largely different BMI groups. Beyond these reports, there is 1 published study documenting the presence of CD4 cells in human fat via immunohistochem-istry,19 and one showing a relative increase in gene expression of FOXP3, a marker for anti-inflammatory Treg cells, in SAT as compared with VAT.13

Indirect support for our finding that adipose tissue Th sub-sets are associated with insulin human resistance is found in 1 published study, demonstrating the presence of both clas-sically activated proinflammatory macrophages and alterna-tively activated anti-inflammatory macrophages in human adipose tissue, with associations between the ratio of pro- to-anti-inflammatory macrophages and insulin resistance as measured by the homeostasis model assessment of insulin resistance.20 Although the signals responsible for macrophage activation in adipose tissue have not been clearly elucidated, we have shown in mice that proinflammatory Th1 cells stimu-late macrophage activation via secretion of IFN-γ.6 Thus, it is conceivable that localized T-cell activation in adipose tissue is a primary event, which results in macrophage recruitment and activation. This is consistent with findings in mice fed a high-fat diet, in which T-cell infiltration into VAT and insulin resistance was observed at 5 weeks but macrophage recruit-ment was not observed until 10 weeks of feeding.19 T-cell secreted cytokines may also directly impair insulin action in target tissues. Inflammatory cytokines (eg, tumor necro-sis factor-α) elaborated by activated T-cells directly impair insulin-mediated glucose uptake via stimulation of IKKB (inhibitor of nuclear factor kappa-B kinase subunit beta) and JNK (c-Jun amino-terminal kinase).1 Other cytokines, such as IL-10, secreted by anti-inflammatory T cells, are associated with enhanced insulin sensitivity and protection from inflam-mation in mice.5 Our results support a protective role for IL-10 and are the first to demonstrate an association between expres-sion of this cytokine in human adipose tissue and systemic insulin sensitivity.

In the context of accumulating data implicating inflamma-tion as causal in the development of atherosclerotic vascular disease and type 2 diabetes mellitus, the potential role of T cells in determining systemic inflammation is of great inter-est. The present results show that T cells in VAT, both CD8 and CD4, adopt a proinflammatory phenotype as compared

with SAT. Furthermore, the frequencies of Th1 and Th2 in VAT are bidirectionally and robustly associated with circulat-ing hsCRP, an inflammatory marker predictive of both type 2 diabetes mellitus and atherosclerotic heart disease. hsCRP is secreted by the liver in response to stimulation by IL-6, a clas-sic proinflammatory cytokine. Given the proximity of VAT to the liver, which is the main source of hsCRP, it is plausible that inflammatory immune cells in VAT provide a particu-larly potent stimulus to hsCRP secretion, via direct cytokine release into the portal vein. In the present study, expression of IL-6, secreted by Th1 and proinflammatory macrophages, was significatntly higher in VAT than in SAT, supporting the hypothesis that proinflammatory immune cells in VAT play a particularly important role in determining hsCRP release by the liver. Whether inflammation in VAT is a stronger deter-minant of systemic inflammation than SAT is an important question, and although prior clinical studies show that abdom-inal and visceral fat mass bear modestly stronger independent associations with plasma hsCRP than total adiposity,21,22 this is the first study to characterize immune cell phenotypes in VAT with respect to systemic inflammation. Additional data sup-porting a role of T cells in determining systemic inflammation are found in an increasing number of studies showing that the balance of pro- to anti-inflammatory T cells is associated with development of atherosclerotic plaque.23

Our study is limited by the relatively small number of presurgical subjects willing to undergo comprehensive insu-lin-resistance testing (SSPG) and the inability to perform comprehensive flow cytometry for all T-cell subsets on every tissue sample. Furthermore, as in most human studies, it is dif-ficult to prove causality. In particular, proving that Th2 cells protect humans from insulin resistance would require selec-tive manipulation of Th2 cells, in vivo, which is not feasible. Considering the present findings together with those from our prior diet-induced obese mice studies, in which transfer of Th2 cells protected against the development of insulin resis-tance,6 the present results are highly suggestive of a similar relationship in humans.

In summary, the present findings extend previous studies in mice and add to the small but growing body of literature in humans implicating T lymphocytes in obesity-related insu-lin resistance and type 2 diabetes mellitus. Not only Th1 and Th2 CD4 cells, as well as IFN-γ+ CD8 T cells, are abundantly present in the VAT and SAT of healthy overweight and obese humans but also the balance of pro- to anti-inflammatory sub-types is associated with systemic inflammation and insulin resistance. The high frequencies of Th1, Th2, and Th17 cells in 2 separate adipose tissue depots are consistent with anti-genic stimulation in these tissues and indicate a widespread effect. Thus, these results provide compelling evidence for a role of the adaptive immune system in systemic inflamma-tion and insulin resistance in overweight or obese humans. Identification of antigens and other key components of this process may ultimately prove useful in the prevention and treatment of obesity-associated metabolic diseases.

Sources of FundingGrant support for this study was provided by National Institutes of Health/National Institute of Digestive Diseases and Diabetes,

by guest on May 17, 2018

http://atvb.ahajournals.org/D

ownloaded from

Page 7: T-Cell Profile in Adipose Tissue Is Associated With ...atvb.ahajournals.org/content/atvbaha/34/12/2637.full.pdfevidence implicates adipose tissue inflammation as a ... tial role of

McLaughlin et al T Cells in Human Adipose Tissue and IR 2643

R01DK080436, R01DK071309, DK096038. Clinical Trials Identifier: NCT00285844.

DisclosuresNone.

References 1. Hotamisligil GS. Inflammation and metabolic disorders. Nature.

2006;444:860–867. 2. Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to

insulin resistance and type 2 diabetes. Nature. 2006;444:840–846. 3. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante

AW Jr Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112:1796–1808.

4. Cinti S, Mitchell G, Barbatelli G, Murano I, Ceresi E, Faloia E, Wang S, Fortier M, Greenberg AS, Obin MS. Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J Lipid Res. 2005;46:2347–2355.

5. Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. 2007;117:175–184.

6. Winer S, Chan Y, Paltsr G, et al. Normalization of obesity-associated insu-lin resistance through immunotherapy. Nature Med. 2009;15:1–10.

7. Harford KA, Reynolds CM, McGillicuddy FC, Roche HM. Fats, inflam-mation and insulin resistance: insights to the role of macrophage and T-cell accumulation in adipose tissue. Proc Nutr Soc. 2011;70:408–417.

8. Nishimura S, Manabe I, Nagasaki M, Eto K, Yamashita H, Ohsugi M, Otsu M, Hara K, Ueki K, Sugiura S, Yoshimura K, Kadowaki T, Nagai R. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med. 2009;15:914–920.

9. Odegaard JI, Ricardo-Gonzalez RR, Goforth MH, Morel CR, Subramanian V, Mukundan L, Red Eagle A, Vats D, Brombacher F, Ferrante AW, Chawla A. Macrophage-specific PPARg controls alternative activation and improves insulin resistance. Nature. 2007;447:1116–1120.

10. Tiemessen MM, Jagger AL, Evans HG, van Herwijnen MJ, John S, Taams LS. CD4+CD25+Foxp3+ regulatory T cells induce alternative activation of human monocytes/macrophages. Proc Natl Acad Sci USA. 2007;104:19446–19451.

11. Hong EG, Ko HJ, Cho YR, et al. Interleukin-10 prevents diet-induced insulin resistance by attenuating macrophage and cytokine response in skeletal muscle. Diabetes. 2009;58:2525–2535.

12. Park-Min KH, Antoniv TT, Ivashkiv LB. Regulation of macrophage phe-notype by long-term exposure to IL-10. Immunobiology. 2005;210:77–86.

13. Feuerer M, Herrero L, Cipolletta D, Naaz A, Wong J, Nayer A, Lee J, Goldfine AB, Benoist C, Shoelson S, Mathis D. Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat Med. 2009;15:930–939.

14. Wu H, Ghosh S, Perrard XD, Feng L, Garcia GE, Perrard JL, Sweeney JF, Peterson LE, Chan L, Smith CW, Ballantyne CM. T-cell accumulation and regulated on activation, normal T cell expressed and secreted upregulation in adipose tissue in obesity. Circulation. 2007;115:1029–1038.

15. Fabbrini E, Cella M, McCartney SA, et al. Association between specific adipose tissue CD4+ T-cell populations and insulin resistance in obese individuals. Gastroenterology. 2013;145:366–374.e1.

16. McLaughlin T, Abbasi F, Lamendola C, Reaven G. Heterogeneity in prev-alence of risk factors for cardiovascular disease and type 2 diabetes in obese individuals: impact of differences in insulin sensitivity. Archives Int Med. 2007;167:642–648.

17. Dalmas E, Venteclef N, Caer C, Poitou C, Cremer I, Aron-Wisnewsky J, Lacroix-Desmazes S, Bayry J, Kaveri SV, Clément K, André S, Guerre-Millo M. T cell-derived IL-22 amplifies IL-1β-driven inflammation in human adipose tissue: relevance to obesity and type 2 diabetes. Diabetes. 2014;63:1966–1977.

18. Bertola A, Ciucci T, Rousseau D, et al. Identification of adipose tissue den-dritic cells correlated with obesity-associated insulin resistance and induc-ing Th17 responses in mice and patients. Diabetes. 2012;61:2238–2247.

19. Kintscher, Hartge M, Hess K, et al. T-lymphocyte infiltration in visceral adipose tissue. Artereoscler Thromb Vasc Biol. 2008;28:1304–1310.

20. Wentworth JM, Naselli G, Brown WA, Doyle L, Phipson B, Smyth GK, Wabitsch M, O’Brien PE, Harrison LC. Pro-inflammatory CD11c+CD206+ adipose tissue macrophages are associated with insulin resistance in human obesity. Diabetes. 2010;59:1648–1656.

21. Pou KM, Massaro JM, Hoffmann U, Vasan RS, Maurovich-Horvat P, Larson MG, Keaney JF Jr, Meigs JB, Lipinska I, Kathiresan S, Murabito JM, O’Donnell CJ, Benjamin EJ, Fox CS. Visceral and subcutaneous adipose tissue volumes are cross-sectionally related to markers of inflam-mation and oxidative stress: the Framingham Heart Study. Circulation. 2007;116:1234–1241.

22. Brooks GC, Blaha MJ, Blumenthal RS. Relation of C-reactive protein to abdominal adiposity. Am J Cardiol. 2010;106:56–61.

23. Lahoute C, Herbin O, Mallat Z, Tedgui A. Adaptive immunity in athero-sclerosis: mechanisms and future therapeutic targets. Nat Rev Cardiol. 2011;8:348–358.

The biological mechanisms linking obesity to insulin resistance are not fully elucidated. It has been suggested that inflammation in adipose tissue may play a role as follows: proinflammatory cytokines and macrophages have been identified in human adipose tissue, and overfeed-ing mice shifts macrophage phenotype in adipose tissue from anti- to proinflammatory. Other types of immune cells may also potentiate localized inflammation and insulin resistance. In mice, the ratio of pro- to anti-inflammatory T-helper cells in adipose tissue increases with weight gain, and blocking their activity prevents obesity-induced insulin resistance. Here, we show that T-helper 1 (proinflammatory) and T-helper 2 (anti-inflammatory) cell frequency in human subcutaneous and visceral adipose tissues correlate with systemic inflammation and insulin resistance. These results extend findings in mice to humans and provide strong support for a role of the adaptive immune system in mediating obesity-induced inflammation and insulin resistance.

Significance

by guest on May 17, 2018

http://atvb.ahajournals.org/D

ownloaded from

Page 8: T-Cell Profile in Adipose Tissue Is Associated With ...atvb.ahajournals.org/content/atvbaha/34/12/2637.full.pdfevidence implicates adipose tissue inflammation as a ... tial role of

EnglemanDaniel Winer, Lorna Tolentino, Okmi Choi, Hong Zhang, Melissa Hui Yen Chng and Edgar Tracey McLaughlin, Li-Fen Liu, Cindy Lamendola, Lei Shen, John Morton, Homero Rivas,

Inflammation in HumansT-Cell Profile in Adipose Tissue Is Associated With Insulin Resistance and Systemic

Print ISSN: 1079-5642. Online ISSN: 1524-4636 Copyright © 2014 American Heart Association, Inc. All rights reserved.

Greenville Avenue, Dallas, TX 75231is published by the American Heart Association, 7272Arteriosclerosis, Thrombosis, and Vascular Biology

doi: 10.1161/ATVBAHA.114.3046362014;

2014;34:2637-2643; originally published online October 23,Arterioscler Thromb Vasc Biol. 

http://atvb.ahajournals.org/content/34/12/2637World Wide Web at:

The online version of this article, along with updated information and services, is located on the

http://atvb.ahajournals.org/content/suppl/2014/10/23/ATVBAHA.114.304636.DC1Data Supplement (unedited) at:

  http://atvb.ahajournals.org//subscriptions/

at: is onlineArteriosclerosis, Thrombosis, and Vascular Biology Information about subscribing to Subscriptions:

  http://www.lww.com/reprints

Information about reprints can be found online at: Reprints: 

document. Question and AnswerPermissions and Rightspage under Services. Further information about this process is available in the

which permission is being requested is located, click Request Permissions in the middle column of the WebCopyright Clearance Center, not the Editorial Office. Once the online version of the published article for

can be obtained via RightsLink, a service of theArteriosclerosis, Thrombosis, and Vascular Biologyin Requests for permissions to reproduce figures, tables, or portions of articles originally publishedPermissions:

by guest on May 17, 2018

http://atvb.ahajournals.org/D

ownloaded from

Page 9: T-Cell Profile in Adipose Tissue Is Associated With ...atvb.ahajournals.org/content/atvbaha/34/12/2637.full.pdfevidence implicates adipose tissue inflammation as a ... tial role of

1

Material and Methods

Subjects: Healthy women and men 35-65 years of age and BMI 25-35 kg/m2 were recruited via

local newspaper advertisements “seeking overweight and moderately-obese healthy

volunteers.” Eligibility required that subjects be nondiabetic, defined as fasting plasma glucose

<126 mg/dL, and free of major organ disease, chronic inflammatory conditions, cancer, active

psychiatric disease, history of liposuction or gastric bypass procedures, pregnancy and

lactation. In addition, subjects were required to be weight-stable for a minimum of three months

and not taking medications known to alter glucose metabolism, immune function, or body

weight. In order to evaluate T-cell subsets (flow cytometry) and their respective cytokines (RT-

PCR) from visceral adipose tissue, subjects were also recruited from the Stanford Bariatric

Surgery Clinic. These subjects, like the others, were required to be healthy and free of diabetes

and major organ disease, although there was no upper limit on BMI (maximum 49.5 kg/m2).

Due to limited tissue sample size and high number of cells required for flow cytometry, in tissue

from subjects with smaller biopsies, flow cytometry was prioritized and thus comparable

subjects with banked frozen tissue, harvested in an identical manner, were used to augment the

sample size for real-time-polymerase chain reaction (RT_PCR) analyses. All subjects gave

written, informed consent, and the protocol was approved by the Stanford University IRB.

Quantification of insulin-mediated glucose uptake: Subjects were admitted to the Stanford

Clinical and Translational Research Unit (CTRU) after a 12 hour overnight fast for measurement

of fasting body weight and height, on standardized scale and stadiometer. Blood was drawn for

measurement of plasma glucose (oximetric method), lipid/lipoproteins (ultracentrifugation), and

hsCRP (bariatric patients). Insulin-mediated glucose uptake was quantified by the modified

insulin suppression test as originally described1, which has been validated with the euglycemic

hyperinsulinemic clamp2. Briefly, after an overnight 12 hour fast, subjects were infused for 180

Page 10: T-Cell Profile in Adipose Tissue Is Associated With ...atvb.ahajournals.org/content/atvbaha/34/12/2637.full.pdfevidence implicates adipose tissue inflammation as a ... tial role of

2

min with octreotide (0.27 µg/m2•min) to suppress endogenous insulin secretion, insulin (25

mU/m2 min), and glucose (240 mg/m2 •min). Blood was drawn at 10-min intervals from 150 to

180 min of the infusion for measurement of plasma glucose and insulin concentrations: the

mean of these values was used as the steady-state plasma insulin (SSPI) and glucose (SSPG)

concentration for each individual. As SSPI concentrations are, by design, similar in all subjects,

the SSPG concentration provides a direct and quantitative measure of the relative ability of

insulin to mediate disposal of an infused glucose load; the higher the SSPG concentration, the

more insulin resistant the individual. Based on a prior study of the distribution of SSPG

concentrations in 449 healthy nondiabetic adults3, we classified volunteers as being insulin

resistant (IR) if their SSPG concentration was in the top tertile, and insulin sensitive (IS) if their

SSPG concentration was in the lowest tertile of the SSPG distribution as previously described.

The use of SSPG tertiles for the purpose of classification is based on prospective studies

showing that individuals with SSPG in the top tertile suffer adverse health effects such as DM2,

cardiovascular disease, and hypertension, whereas those in the lowest SSPG tertile do not4,5.

Laboratory analysts were blinded as to insulin resistance status of the subjects. High-sensitivity

C-reactive protein (hsCRP) was quantified on fasting plasma samples by immunoassay.

Human IL6 measurement

Human inflammatory cytokine kit (BD bioscience Cytometric Bead Array, CBA) was used to

measure IL6 levels in ETDA-treated plasma from human subjects. 10uL samples were run in

duplicate. CBA assays provide a method of capturing a soluble analyte or set of analytes with

beads of known size and fluorescence. Fluorescence was measured using flow cytometry.

Adipose tissue biopsies: After an overnight fast, under sterile conditions and local anesthesia,

nonbariatric surgery subjects underwent superficial (just under the skin) subcutaneous

periumbilical adipose tissue biopsies as previously described6. For bariatric surgery patients,

Page 11: T-Cell Profile in Adipose Tissue Is Associated With ...atvb.ahajournals.org/content/atvbaha/34/12/2637.full.pdfevidence implicates adipose tissue inflammation as a ... tial role of

3

superficial SAT from the periumbilical area and VAT from the greater omentum were obtained in

a standardized manner at the outset of the bariatric surgery procedure and immediately

processed by laboratory personnel. For both surgical and nonsurgical patients, a portion of

adipose tissue was flash frozen on liquid nitrogen for preparation of cDNA. Isolation of

adipocytes and stromal-vascular cells from remaining tissue was performed via collagenase

digestion using a modification of the original protocol of Rodbell7. Sterile harvested tissue was

immediately placed in sterile PBS with 2% bovine serum albumin until separation was initiated.

Mechanically minced adipose tissue was digested for 30 min at 37°C in a shaking water bath

with collagenase I, 1mg/mL in 0.1M HEPES, 0.12M NaCl, 0.05M KCL, 0.005M glucose, 1.5%

wt/vol BSA, 1mM CaCl2, 2H2O, pH7.4. The cell suspension was then filtered through a 500-um

nylon mesh and centrifuged for 5 min at 500RPM at room temperature. Isolated cells were

washed and mature fat cells removed by flotation. The resulting cell pellet was collected after

centrifugation at 1200 RPM for 5 minutes and then incubated in erythrocyte lysis buffer

(Invitrogen) for 10 min at 37 C. PBS with 2% BSA was added to the cell suspension following

erythrocyte lysis procedure.

Flow cytometry: Single cell suspensions of mononuclear cells from fresh adipose tissue and

heparinized whole blood were analyzed by multicolor flow cytometry for the frequency and

activation state of CD4 T cell subsets as previously described8. These experiments utilized

fluorescently labeled monoclonal antibodies (mAbs) specific for CD2, CD4, CD8, CD14, CD16,

CD19, CD56, CD45, IL-13, IL-17, and IFN-γ, along with the appropriate isotype-matched control

Abs. Cells were analyzed directly or, for intracellular cytokine detection, stimulated with PMA

(10 ng/ml) and Ionomycin (1µg/ml) for 4 hours with addition of Brefeldin A (10 µg/ml) 1-hour

post stimulation. Subsets of interest included Th1 (IFN+), Th2 (IL-13+), Th17 (IL-17+) and Treg

(FoxP3+), which were expressed as % CD4 cells, and total CD4 and CD8 T cells, expressed as

% total T cells. Details of the gating procedure are shown in Supplemental Figure 1.

Page 12: T-Cell Profile in Adipose Tissue Is Associated With ...atvb.ahajournals.org/content/atvbaha/34/12/2637.full.pdfevidence implicates adipose tissue inflammation as a ... tial role of

4

Stimulated cells were subsequently permeabilized with Cytofix/Cytoperm and analyzed by flow

cytometry with BD LSRII and FACSDiva software. To identify Tregs, cells surface-labeled cells

were resuspended with 1 ml of Fix/perm buffer (1x) and incubated at room temperature for 20

min. After washing twice with MACS buffer, cells were incubated with FoxP3 perm buffer at

room temperature for 15 minutes. The supernatant was discarded and FoxP3 Perm buffer (100

ul) was added to the cells prior to staining with intracellular FoxP3 Pacific Blue antibody

(Biolegend), for an additional 30 min. Cells were washed and resuspended with MACS buffer

prior to flow cytometry analysis. The cells were kept in the dark during the process. Although in

the current study identification of Th2 cells was based on detection of intracellular IL-13 in CD4

T cells, under the assay conditions used there is >95% overlap between IL-13 and IL-4

producing cells. All fluorochrome-conjugated antibodies and reagents were purchased from

either BD Biosciences or Biolegend.

Gene expression: Total RNA was extracted from frozen adipose tissue using TriZol (Invitrogen,

Carlsbad, CA) and the Adipose Tissue RNAeasy kit (Qiagen, Valencia, CA) according to the

manufacturer’s instructions. cDNA was synthesized from total RNA using High Capacity RNA-

to-cDNA kit (Applied Biosystems, Carlsbad, CA). A threshold cycle (Ct value) was obtained from

each amplification curve, and a Δ Ct value calculated by subtracting the Ct value for 18S

ribosomal RNA from the Ct value for each sample. A ΔΔCt value was then calculated by

subtracting the Δ Ct value of a SAT adipocyte sample (control). Gene expression of cytokines

reflective of pro-inflammatory Th1 and Th17 activation (IL-6, IFNγ and IL-17) as well as anti-

inflammatory IL-10 were quantified, using Taqman primers/probes (Applied Biosystems). Fold

changes vs control were determined using the comparative threshold method (2−ΔΔCt)9.

Immunohistochemistry: Fresh SAT and VAT tissue samples were fixed in 10% formalin then

paraffin embedded by Histo-Tec laboratory (Histo-Tec, Hayward CA). Sections were

Page 13: T-Cell Profile in Adipose Tissue Is Associated With ...atvb.ahajournals.org/content/atvbaha/34/12/2637.full.pdfevidence implicates adipose tissue inflammation as a ... tial role of

5

deparaffinized in 3 changes of xylene followed by graded alcohols. Antigen retrieval was

performed using pH 6.0 Diva universal retrieval solution (Biocare Medical) inside a pressure

cooker (Biocare Medical). Endogenous peroxidase activity was quenched using Peroxidazed 1

(Biocare Medical). Sections were blocked with serum-free protein blocker (Dako). The primary

antibodies used were: mouse monoclonal anti-CD4 (clone: 4B12, 1:10, Vector), rabbit

monoclonal anti-CD8 (clone: EP1150Y, 1:100, Abcam). For detection, MACH2 Double Stain 2

(Biocare Medical) was used. Color development was performed with freshly prepared 3,3′

Diaminobenzidine (DAB) solution and Ferangi Blue solution (Biocare Medical) before mounting

with Faramount Mounting Medium (Dako).

Statistical analysis: Data are expressed as mean + standard deviation. Normality of data was

tested via quantile plots. Non-normally distributed data (PBMC Th2 and hsCRP) were log-

transformed for statistical tests. Comparison of gene expression and T-cell data between

adipose tissue depots and peripheral blood was done utilizing paired Student’s t-test. Pearson’s

correlations were used to determine univariate associations between T-cell variables in tissue

and blood with insulin resistance (SSPG). Linear regression with adjustment for BMI was used

to measure associations between T-cell frequency and IL-10 expression and insulin resistance

(SSPG), as well as between Th frequency and plasma cytokines. P< 0.05 was considered

statistically significant.

References

1. Pei D, Jones CN, Bhargava R, Chen YD, Reaven GM. Evaluation of octreotide of assess

insulin-mediated glucose disposal by the insulin suppression test. Diabetologia 2004,

37:843-5.

Page 14: T-Cell Profile in Adipose Tissue Is Associated With ...atvb.ahajournals.org/content/atvbaha/34/12/2637.full.pdfevidence implicates adipose tissue inflammation as a ... tial role of

6

2. Greenfield M, Loberne L, Kraemer F, Tobey T, Reaven G. Assessment of insulin resistance

with the insulin suppression test and euglycemic clamp. Diabetes 1981, 30:387-92.

3. Yeni-Komshian H, Carantoni M, Abbasi F, Reaven GM. Relationship between several

surrogate estimates of insulin resistance and quantification of insulin-mediated glucose

disposal in 490 healthy nondiabetic volunteers. Diabetes Care 2000; 23:171-175

4. Yip J, Gacchini FS, Reaven GM. Resistance to insulin-mediated glucose disposal as a

predictor of cardiovascular disease. J Clin Endocrinol Metab 1998, 83:2773-2777.

5. Facchini FS, Hua N, Abbasi F, Reaven GM. Insulin resistance as a predictor of age-related-

diseases. J Clin Endocrinol Metab 2001, 86:3574-3578.

6. McLaughlin T, Sherman A, Tsao P, Gonzalez O, Yee G, et al. Enhanced proportion of small

adipose cells in insulin-resistant vs insulin -sensitive obese individuals implicates impaired

adipogenesis. Diabetologia 2007, 50:1707-1715.

7. Rodbell M. Metabolism of isolated fat cells. Effects of hormones on glucose metabolism and

lipolysis. J Biol Chem 1964, 239:375-380.

8. Picker LJ, Singh MK, Zdraveski Z, Treer JR, Waldrop SL, Bergstresser PR, Maino VC.

Direct Demonstration of Cytokine Synthesis Heterogeneity Among Human Memory/Effector

T Cells by Flow Cytometry. Blood 1995:86: 1408-1419.

9. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time

quantitativePCR and the 2(-Delta Delta C(T)) Method 1. Methods 2001, 25: 402-408.

Page 15: T-Cell Profile in Adipose Tissue Is Associated With ...atvb.ahajournals.org/content/atvbaha/34/12/2637.full.pdfevidence implicates adipose tissue inflammation as a ... tial role of

1

Supplement Material

Supplementary Table I. Subject characteristics (mean + SD)

Variable Nonbariatric

Subjects for T Cells

(n=13)

Bariatric Subjects

for T Cells

(n=10)

Nonbariatric

Subjects for rtPCR

(n=20*)

Bariatric Subjects

for rtPCR

(n=10)

Age (yrs) 54.2 + 4.8 42.9 + 12.5 56 + 9.3 45.3 + 12.6

Sex (F/M) 8/5 7/3 update 19/1 6/4

Race (C/H/B/A) 8/1/3/0 5/3/2/0 13/2/5/0 5/4/1/0

BMI (kg/m2) 29.2 + 2.1 45.3 + 3.4 31.7 + 4.6 46.2 + 8.6

SSPG (mg/dL) 127 + 52 231 + 88 166 + 80 NA

*includes 6 subjects from the T cell cohort and 14 separate subjects for a total of 27 nonbariatric subjects

rtPCR: real time polymerase chain reaction

C: Caucasian; H: Hispanic; B: Black; A: Asian

Page 16: T-Cell Profile in Adipose Tissue Is Associated With ...atvb.ahajournals.org/content/atvbaha/34/12/2637.full.pdfevidence implicates adipose tissue inflammation as a ... tial role of

2

A

B

C

Page 17: T-Cell Profile in Adipose Tissue Is Associated With ...atvb.ahajournals.org/content/atvbaha/34/12/2637.full.pdfevidence implicates adipose tissue inflammation as a ... tial role of

3

Supplementary Figure I. Gating strategy to separate T cell subsets via flow cytometry performed on human adipose

tissue and blood. Panel A: T helper cells: After gating out doublet cells, the CD45+ lymphocyte population is selected

and non-viable cells are excluded. CD2+ T cells are separated from other lineages. Side scatter and CD8 expression

are then used to identify CD4 and CD8 populations. Positive selection of CD4 T cells is avoided due to down

modulation of the CD4 molecule upon activation with PMA/Ionomycin. IFNg , IL13 ,and IL17 are plotted against CD4

to determine TH1, TH2, TH17 expression, respectively.

Panel B: Plots displaying IFNγ, IL13 and IL17 expression of CD4 cells before and after stimulation with

PMA/Ionomycin.

Panel C: Regulatory T cells: After gating out doublets, the CD45+ lymphocyte population is selected and non-viable

cells are excluded. CD2+ cells are excluded from other lineages. Side scatter and CD8 expression are then used to

identify CD4 and CD8 population. CD4+ cells expressing FoxP3 are then identified as regulatory T cells.

Supplementary Figure II. Relationship between plasma interleukin-6 concentrations in association with human

subcutaneous adipose tissue (SAT) Th 1 frequency.