17
Mawadda Alnaeeli, 1 Bruce M. Raaka, 2 Oksana Gavrilova, 3 Ruifeng Teng, 3 Tatyana Chanturiya, 3 and Constance Tom Noguchi 1 Erythropoietin Signaling: A Novel Regulator of White Adipose Tissue Inammation During Diet-Induced Obesity Diabetes 2014;63:24152431 | DOI: 10.2337/db13-0883 Obesity-induced white adipose tissue (WAT) inamma- tion and insulin resistance are associated with mac- rophage (Mv) inltration and phenotypic shift from anti-inammatoryM2-like to predominantly proinam- matoryM1-like cells. Erythropoietin (EPO), a glycoprotein hormone indispensable for erythropoiesis, has biologi- cal activities that extend to nonerythroid tissues, includ- ing antiapoptotic and anti-inammatory effects. Using comprehensive in vivo and in vitro analyses in mice, EPO treatment inhibited WAT inammation, normalized insulin sensitivity, and reduced glucose intolerance. We investigated EPO receptor (EPO-R) expression in WAT and characterized the role of its signaling during obesity-induced inammation. Remarkably, and prior to any detectable changes in body weight or composi- tion, EPO treatment reduced M1-like Mv and increased M2-like Mv in WAT, while decreasing inammatory monocytes. These anti-inammatory effects were found to be driven, at least in part, by direct EPO-R response in Mv via Stat3 activation, where EPO effects on M2 but not M1 Mv required interleukin-4 receptor/Stat6. Using obese ΔEpoR mice with EPO-R restricted to erythroid cells, we demonstrated an anti-inammatory role for en- dogenous EPO. Collectively, our ndings identify EPO-R signaling as a novel regulator of WAT inammation, extending its nonerythroid activity to encompass effects on both Mv inltration and subset composition in WAT. Macrophage (Mv) inltration to white adipose tissue (WAT) during obesity marks a state of chronic inammation, an important cause of obesity-induced insulin resistance and glucose intolerance (14). This chronic inammatory re- sponse impacts type 2 diabetes pathogenesis (1,4) and asso- ciates with a shift in Mv population from alternatively activated F4/80 + MGL-1 + (anti-inammatory) M2 to predom- inantly classically activated F4/80 + MGL-1 2 (proinamma- tory) M1 (58) in association with recruitment from circulating inammatoryLy6C hi CCR2 + monocytes to Mv clusters in WAT via CCL2/CCR2 axis and not to the con- version of resident M2 Mv to M1 (1,3,5,6,810). Mv inltration and activation state inuence inammation-induced insulin resistance and glucose in- tolerance during diet-induced obesity (DIO) (1113). It is suggested that Mv may be an initiator in insulin-resistant states and that, with their precursors, may contribute to propagation of insulin resistance (1117). Mv inltration to WAT requires and kinetically follows CD8 + T lympho- cytes recruitment (18). Obesity-induced inammation and immune cell inltration elevate cytokines and chemokines such as tumor necrosis factor (TNF)-a and CCL2, sys- temically and locally in WAT, where cells of the stromal vascular fraction (SVF), including Mv, are known to be the main producers, particularly in visceral fat depots (1,3,4,1921). Signaling of such inammatory mediators 1 Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 2 Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 3 Mouse Metabolism Core Facility, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD Corresponding author: Constance Tom Noguchi, [email protected]. Received 5 June 2013 and accepted 5 March 2014. This article contains Supplementary Data online at http://diabetes .diabetesjournals.org/lookup/suppl/doi:10.2337/db13-0883/-/DC1. M.A. is currently afliated with the Department of Biological Sciences, Ohio University, Zanesville, OH, and the Diabetes Institute at Ohio University, Athens, OH. © 2014 by the American Diabetes Association. See http://creativecommons.org /licenses/by-nc-nd/3.0/ for details. See accompanying article, p. 2229. Diabetes Volume 63, July 2014 2415 PATHOPHYSIOLOGY

Erythropoietin Signaling: A Novel Regulator of White ...diabetes.diabetesjournals.org/content/diabetes/63/7/2415.full.pdf · hormone indispensable for erythropoiesis, has biologi-cal

  • Upload
    buikiet

  • View
    213

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Erythropoietin Signaling: A Novel Regulator of White ...diabetes.diabetesjournals.org/content/diabetes/63/7/2415.full.pdf · hormone indispensable for erythropoiesis, has biologi-cal

Mawadda Alnaeeli,1 Bruce M. Raaka,2 Oksana Gavrilova,3 Ruifeng Teng,3 Tatyana Chanturiya,3 andConstance Tom Noguchi1

Erythropoietin Signaling:A Novel Regulator of WhiteAdipose Tissue InflammationDuring Diet-Induced ObesityDiabetes 2014;63:2415–2431 | DOI: 10.2337/db13-0883

Obesity-induced white adipose tissue (WAT) inflamma-tion and insulin resistance are associated with mac-rophage (Mv) infiltration and phenotypic shift from“anti-inflammatory” M2-like to predominantly “proinflam-matory”M1-like cells. Erythropoietin (EPO), a glycoproteinhormone indispensable for erythropoiesis, has biologi-cal activities that extend to nonerythroid tissues, includ-ing antiapoptotic and anti-inflammatory effects. Usingcomprehensive in vivo and in vitro analyses in mice,EPO treatment inhibited WAT inflammation, normalizedinsulin sensitivity, and reduced glucose intolerance. Weinvestigated EPO receptor (EPO-R) expression in WATand characterized the role of its signaling duringobesity-induced inflammation. Remarkably, and priorto any detectable changes in body weight or composi-tion, EPO treatment reduced M1-like Mv and increasedM2-like Mv in WAT, while decreasing inflammatorymonocytes. These anti-inflammatory effects were foundto be driven, at least in part, by direct EPO-R response inMv via Stat3 activation, where EPO effects on M2 butnot M1 Mv required interleukin-4 receptor/Stat6. Usingobese ΔEpoR mice with EPO-R restricted to erythroidcells, we demonstrated an anti-inflammatory role for en-dogenous EPO. Collectively, our findings identify EPO-Rsignaling as a novel regulator of WAT inflammation,extending its nonerythroid activity to encompass effectson both Mv infiltration and subset composition in WAT.

Macrophage (Mv) infiltration to white adipose tissue (WAT)during obesity marks a state of chronic inflammation, animportant cause of obesity-induced insulin resistance andglucose intolerance (1–4). This chronic inflammatory re-sponse impacts type 2 diabetes pathogenesis (1,4) and asso-ciates with a shift in Mv population from alternativelyactivated F4/80+MGL-1+ (anti-inflammatory) M2 to predom-inantly classically activated F4/80+MGL-12 (proinflamma-tory) M1 (5–8) in association with recruitment fromcirculating “inflammatory” Ly6ChiCCR2+ monocytes to Mvclusters in WAT via CCL2/CCR2 axis and not to the con-version of resident M2 Mv to M1 (1,3,5,6,8–10).

Mv infiltration and activation state influenceinflammation-induced insulin resistance and glucose in-tolerance during diet-induced obesity (DIO) (11–13). It issuggested that Mv may be an initiator in insulin-resistantstates and that, with their precursors, may contribute topropagation of insulin resistance (11–17). Mv infiltrationto WAT requires and kinetically follows CD8+ T lympho-cytes recruitment (18). Obesity-induced inflammation andimmune cell infiltration elevate cytokines and chemokinessuch as tumor necrosis factor (TNF)-a and CCL2, sys-temically and locally in WAT, where cells of the stromalvascular fraction (SVF), including Mv, are known to bethe main producers, particularly in visceral fat depots(1,3,4,19–21). Signaling of such inflammatory mediators

1Molecular Medicine Branch, National Institute of Diabetes and Digestive andKidney Diseases, National Institutes of Health, Bethesda, MD2Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetesand Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD3Mouse Metabolism Core Facility, National Institute of Diabetes and Digestive andKidney Diseases, National Institutes of Health, Bethesda, MD

Corresponding author: Constance Tom Noguchi, [email protected].

Received 5 June 2013 and accepted 5 March 2014.

This article contains Supplementary Data online at http://diabetes.diabetesjournals.org/lookup/suppl/doi:10.2337/db13-0883/-/DC1.

M.A. is currently affiliated with the Department of Biological Sciences, OhioUniversity, Zanesville, OH, and the Diabetes Institute at Ohio University,Athens, OH.

© 2014 by the American Diabetes Association. See http://creativecommons.org/licenses/by-nc-nd/3.0/ for details.

See accompanying article, p. 2229.

Diabetes Volume 63, July 2014 2415

PATHOPHYSIO

LOGY

Page 2: Erythropoietin Signaling: A Novel Regulator of White ...diabetes.diabetesjournals.org/content/diabetes/63/7/2415.full.pdf · hormone indispensable for erythropoiesis, has biologi-cal

is adversely implicated in the impairment of systemic glu-cose metabolism (1,5).

Erythropoietin (EPO) is a glycoprotein hormone in-duced by hypoxia and necessary for erythrocyte pro-duction (22–24). EPO is used for treatment of anemia inchronic kidney disease, including type 2 diabetic patients(25). Its biological activity extends beyond regulatingerythropoiesis, and the nonerythroid expression of EPOreceptor (EPO-R) has been reported (26–31). EPO wasreported to reduce Mv infiltration and inhibit inflamma-tion (32). Although, proposed to occur via antiapoptoticrather than “direct” anti-inflammatory effects on cells ofthe immune system (32), it can also occur directly viainhibiting Mv and/or activating immune suppressivelymphocytes (33,34).

Early treatment of mice with exogenous EPO at theonset of high-fat diet (HFD) feeding or EPO transgenicoverexpression halts body weight and fat mass gain andimproves glucose tolerance (35–38). Using ΔEpoR mice withEPO-R restricted to erythroid tissue (39), we previouslyshowed EPO-R absence in WAT contributes directly to obe-sity and glucose intolerance on normal chow (36). EPOprotects against diabetes through direct effects on pancre-atic b-cells in mouse models of types 1 and 2 diabetes(36,37,40). It remains unknown whether anti-inflammatoryEPO effects in WAT contribute to EPO effects on the pre-diabetic state during obesity.

In this study, we hypothesized that EPO/EPO-R signal-ing can attenuate obesity-induced WAT inflammation. Ourfindings identify a novel role for EPO in regulating in-flammatory monocyte recruitment and Mv infiltrationand activation during DIO.

RESEARCH DESIGN AND METHODS

Animals and Animal CareWild-type (WT) male C57BL/6 mice were obtained fromNational Cancer Institute Animal Production Program(Frederick, MD). Mice with EPO-R expression restrictedto hematopoietic tissue (ΔEpoR) were provided byMasayuki Yamamoto (Tohoku University, Japan). EPO-Rexpression in ΔEpoR mice arises from transgene expres-sion of EpoR cDNA driven by the erythroid specificenhancer/promoter of GATA-1 on C57BL/6 EpoR2/2

background (29). Male Stat62/2 (Stat6tm1Gru) and in-terleukin (IL)-42/2 (ll4tm1Nnt) mice on C57BL/6 back-ground and their age-matched WT controls were obtainedfrom The Jackson Laboratory (JAX mice) (Bar Harbor,ME). Mice were fed control diet (10 kcal% fat) or, toinduce DIO, HFD containing 60 kcal% fat (high fat,5,240 kcal/kg, 34.9% crude fat; Research Diets Inc.,New Brunswick, NJ) starting at 6–8 weeks of age for 12weeks. During the last 2 weeks of the study period, obese,insulin-resistant, and glucose-intolerant mice were subcu-taneously injected with 1,000 units/kg EPO (EPO dosewas chosen based on our previous titration studies [35])or saline as a vehicle control, every 48 h while continuingHFD feeding. At the end of week 12, the effects of 2

weeks EPO treatment on several metabolic and inflam-matory parameters were assessed as described below. Ani-mals were housed under specific pathogen-free conditionsand maintained in a thermostated environment undera 12-h light/dark cycle with free access to food and water.All animal protocols were conducted under the NationalInstitutes of Health guidelines and approved by the insti-tute’s Animal Care and Use Committee.

Reagents and AntibodiesReagents and antibodies include anti-F4/80 and anti-Ly6Cantibodies (eBioscience); anti-MGL1/CD301 and anti-CCR2(R&D Systems); Fc-block and anti-CD11c antibodies (BDBiosciences); rat anti-mouse Ki-67 antibody (BioLegend)used in combination with fluorescein isothiocyanate–conjugated goat anti-rat F(ab)2 (Santa Cruz BiotechnologyInc.); bromodeoxyuridine (BrdU; Sigma-Aldrich) and fluores-cein isothiocyanate BrdU flow detection kit (BD Pharmingen);rabbit anti-mouse phospho (p)-Stat5 (Tyr694; C71E5) andp-Stat3 (Tyr705; D3A7) monoclonal antibodies (Cell Signal-ing Technology); mouse anti-mouse phopho-Stat6 (Tyr641;BD Biosciences); recombinant human Epoetin a or Epogen(EPO) (Amgen); Vectashield mounting media with DAPI(Vector Laboratories); and recombinant mouse cytokinesGM-CSF, IL-4, and IL-10 (PeproTech).

Body Weight and Composition MeasurementsBody weight was measured weekly from day 0 to week 12.Body composition analysis to determine fat and lean bodymass was also performed using EchoMRI-100 system(Echo Medical Systems).

Hematocrit MeasurementHematocrit was determined for saline- and EPO-treatedanimals after centrifugation of blood containing heparincoated capillary tubes, using VIN microcapillary tubereader (Veterinary Information Network Bookstore).

Metabolic EvaluationGlucose tolerance tests (GTTs) were performed upon in-traperitoneal injection with 1 g/kg dextrose. Insulin tolerancetests (ITTs) were performed after injection with 1 unit/kginsulin. Total blood and serum glucose measurements weremade using AlphaTRAK glucometer (Abbott Animal Health).Serum insulin levels were measured using Sensitive RatInsulin Radioimmunoassay (Millipore). Serum leptinlevels were determined by ELISA (Millipore). Euglycemic–hyperinsulinemic clamp studies were performed in re-strained mice fasted for 16 h as described previously (41).

SVF Cell ExtractionSVF cells were extracted as previously described (18).Briefly, perigonadal WAT were digested by collagenasetype 2 (Worthington) at 37°C for 50 min. WAT cell sus-pensions were spun at 500g for 5 min to separate floatingadipocytes from SVF pellet.

Gene Expression AnalysisTotal RNA was extracted from cells or tissues usingTRIzol (Invitrogen) and treated with DNase I (Fermentas)

2416 Erythropoietin and Obesity-Induced Inflammation Diabetes Volume 63, July 2014

Page 3: Erythropoietin Signaling: A Novel Regulator of White ...diabetes.diabetesjournals.org/content/diabetes/63/7/2415.full.pdf · hormone indispensable for erythropoiesis, has biologi-cal

according to manufacturers’ protocols. Two micrograms oftotal RNA was then reverse transcribed using MultiScribeReverse Transcriptase for quantitative real-time PCR(qRT-PCR) assays using ABI 7900HT cycler (Applied Bio-systems). For EPO-R gene expression analysis, mRNAwas quantified using TaqMan qRT-PCR, with 16S andb-actin as controls. Relative changes in gene expressionlevels of inflammatory cytokines, chemokines, and recep-tors, in SVF of epididymal adipose tissue, were analyzedusing mouse inflammatory cytokines and receptors RT2

profiler PCR (SABiosciences, Qiagen). Briefly, cDNA wassynthesized using RT2 first strand reaction, and PCR wasperformed using RT2 PCR master mix (SABiosciences,Qiagen). Four to six replicate 96-well plates were in-cluded per group; data analysis was performed usingweb-based PCR Array Data Analysis Software version3.4 (SABiosciences, Qiagen). Changes in Mv subtype sig-nature gene expression levels M1 (inducible nitric oxidesynthase [iNOS] and IL-1b) and M2 (Fizz-1, Arg-1,peroxisome proliferator–activated receptor [Ppar]-g)were assessed in SVF cells using sybergreen gene expres-sion assays (Fermentas). Relative gene expression levelswere determined by normalization to b-actin expressionlevels using the D–D Ct method. Primer and probesequences used are listed in Supplementary Table 1.

WAT and Serum Cytokine and Chemokine ProteinMeasurementsCCL2 levels were analyzed in perigonadal WAT tissue andserum; those for TNF-a, IL-10, and IL-4 were analyzed inserum by ELISA according to the manufacturer’s protocol(PeproTech).

Cell Sorting and PurificationWAT Mv were sorted by separation of F4/80+ (Mv) andF4/802 (non-Mv) fractions of stromal vascular cells us-ing FACSAria fluorescent cell sorter and FACSDiva soft-ware (BD Biosciences). Sorted WAT Mv were used forqRT-PCR analysis of EPO-R expression levels or culturedto assess Stat5, Stat3, and Stat6 phosphorylation.

EPO Treatment of Sorted WAT Mv In VitroWAT Mv, fluorescence-activated cell sorter (FACS) sortedfrom SVF of obese mice (after 12 weeks of HFD feeding)was cultured in saline or EPO (5 units/mL)-coated 48-wellplates at a density of 2 3 105 per well for 15 min thenfixed, permeabilized, and tested for Stat3, Stat5a/b, andStat6 phosphorylation by phosphoflow. Stimulation for15 min with GM-CSF, IL-4, and IL-10 were used as pos-itive controls for Stat5, Stat6, and Stat3 phosphorylation,respectively. In some studies, the sorted WAT Mv werecultured with EPO (5 units/mL) for 24 h, after which theirTNF-a, iNOS, and IL-10 mRNA levels were quantifiedusing sybergreen qRT-PCR.

Flow Cytometry and PhosphoflowSVF of perigonadal WAT was analyzed by flow cytometry aspreviously described (8). Circulating inflammatory mono-cytes were quantified using Ly6C and CCR2 expression as

previously described (42). Phosphoflow and staining forp-Stat3, p-Stat5a/b, and p-Stat6 were performed as pre-viously described (43). Cell proliferation was assessed usingKi-67 detection and BrdU labeling. In vivo labeling withBrdU was achieved by supplementing drinking water with0.8 mg/mL BrdU, starting 48 h prior to the first EPO in-jection during weeks 11 and 12 of HFD, for 9 consecutivedays followed by a changeover to normal water untilthe end of week 12. FACSCalibur and CellQuest (BD Bio-sciences) were used for flow cytometric and phosphoflowanalyses.

Histology and MicroscopyFrozen sections of perigonadal fat pads were preparedafter dissection and fixation in 10% formalin. Hematox-ylin and eosin (H&E)-stained sections were imagedfor crown-like structures. Immunofluorescence was per-formed for Mv detection by incubation en block withdirectly conjugated primary anti-F4/80 antibodies. All im-aging analyses were performed on OLYMPUS IX70inverted microscope (Optical Elements Corporation). Im-age capture was performed using SPOTFlex camera andSPOT Basic imaging software (Diagnostic InstrumentsInc.), using 15 sections per group (n = 5 mice per group).

Statistical AnalysisStatistical comparisons were performed using Studentt test, with P values ,0.05 defined as statistically signif-icant. Results are presented as mean 6 SEM.

RESULTS

EPO Treatment Regimen Has No Effect on BodyWeight or Fat Mass During DIOEPO treatment of obese mice reduces body weight andfat mass after 3–4 weeks (35). Considering that bodyweight and fat mass changes influence WAT inflamma-tion and Mv infiltration, affecting glucose tolerance andinsulin sensitivity (1,2,44), we chose short-term EPOtreatment regimen (,3 weeks) after obesity induction.Obese, insulin-resistant, glucose-intolerant mice, fed anHFD for 12 weeks, were EPO treated during the last 2weeks of the study with no detectable effects on bodyweight, body weight gain, fat mass, perigonadal fatweight, or serum leptin levels (Fig. 1A–E). Consistentwith previous reports (35), prolonged EPO adminis-tration ($3 weeks) reduced body weight (Fig. 1F). Allsubsequent studies were conducted using the 2-weekregimen of EPO treatment.

EPO Treatment Attenuates Insulin Resistance andGlucose Intolerance During DIOWe investigated whether EPO treatment can ameliorateexistent systemic insulin resistance and glucose intoleranceassociated with DIO using ITT and GTT. EPO improved both(Fig. 2A and B), with striking reduction in fasting bloodglucose levels (Fig. 2C) and impressive effects on glucosetolerance (Fig. 2B). EPO increased hematocrit (Fig. 2D); how-ever, serum glucose (Fig. 2E) and serum insulin (Fig. 2F) werealso reduced, showing that total blood glucose reduction is

diabetes.diabetesjournals.org Alnaeeli and Associates 2417

Page 4: Erythropoietin Signaling: A Novel Regulator of White ...diabetes.diabetesjournals.org/content/diabetes/63/7/2415.full.pdf · hormone indispensable for erythropoiesis, has biologi-cal

not the result of increased packed red cell volume. To exam-ine insulin sensitivity of individual tissues, we performedeuglycemic–hyperinsulinemic clamp studies. EPO-treatedmice required higher glucose infusion rate (GIR) to maintainblood glucose during insulin infusion and exhibited higherwhole body glucose disposal and glycogen synthesis ratecompared with controls (Fig. 2G and H). Although thesedifferences have not reached statistical significance, EPOsignificantly increased 2-deoxyglucose uptake in skeletal mus-cle but not WAT, brown adipose tissue, or heart (Fig. 2I andJ). EPO had no effect on basal or clamp endogenous glucoseproduction (Fig. 2H), suggesting no changes in liver insulinsensitivity. Taken together, these data demonstrate that EPOtreatment, which had no effect on body weight, improved

whole body insulin sensitivity primarily by enhancing glucoseuptake in the skeletal muscles.

EPO Treatment Inhibits WAT Inflammation andReduces Mv InfiltrationUnlike WAT with high EPO-R expression in both adipocytesand SVF, EPO-R is not detectable in skeletal muscle (Fig. 3Aand B), indicating that EPO indirectly stimulates glucoseuptake in skeletal muscle. Although EPO had no effect onWAT insulin sensitivity, we noticed that it dramatically im-proved inflammation within adipose tissue, as indicatedby the lack of crown structure around adipocytes in EPO-treated mice (Fig. 3C and D). This novel observation promptedus to study the role of EPO signaling in obesity-induced

Figure 1—A 2-week EPO treatment regimen does not affect body weight or fat mass. WT C57BL/6 male mice were fed an HFD for 12weeks and treated with or without EPO (1,000 units/kg) for the final 2 weeks of the study. Lean mice + saline and obese mice + saline wereused as negative and vehicle controls, respectively. Final body weight (A), body weight gain (B), fat mass (C ), perigonadal fat mass (D), andserum leptin (E) were measured for all three groups at the end of week 12. F: WT C57BL/6 mice with obesity induced by HFD feeding for 13weeks were treated with or without EPO (1,000 units/kg) every 48 h during the final 3 weeks of the study. Lean mice + saline were used asnegative controls. Results are shown as mean 6 SEM for n = 8 mice per group, representative of three independent experiments withsimilar results. *P < 0.05.

2418 Erythropoietin and Obesity-Induced Inflammation Diabetes Volume 63, July 2014

Page 5: Erythropoietin Signaling: A Novel Regulator of White ...diabetes.diabetesjournals.org/content/diabetes/63/7/2415.full.pdf · hormone indispensable for erythropoiesis, has biologi-cal

inflammation. We performed comprehensive gene expres-sion profiling of cytokines and chemokines in SVF. EPOreduced Ccl1, Ccl2, Ccl3, and Ccl22 expression levels butnot IL-10 (Fig. 3E). Protein analysis showed that EPOtreatment reduced TNF-a—consistent with the role ofTNF-a during obesity-induced insulin resistance (45)—increased IL-10 (Fig. 3F), and downregulated CCL1,CCL2, CCL3, and CCL22 levels (data not shown).

Mv are responsible for the majority of EPO-R expres-sion in SVF (Fig. 3G), and EPO treatment reduced total Mvpercentage and number by ;11.2 and ;30%, respectively(Fig. 3H and I), supporting an inhibitory effect for EPO onWAT inflammation. Mv are the main responders in SVF,as T-cell and B-cell percentage showed no detectablechanges after EPO treatment (Supplementary Table 2). Con-sistent with results showing Mv as predominant EPO-R

Figure 2—EPO treatment attenuates insulin resistance and glucose intolerance during DIO. WT C57BL/6 mice with obesity induced byHFD feeding for 12 weeks were treated with or without EPO (1,000 units/kg) every 48 h during the final 2 weeks of the study. Lean mice +saline were used as negative controls. A: For ITT, glucose levels were measured after intraperitoneal injection of 1 unit/kg insulin. B: ForGTT, glucose levels were measured after intraperitoneal injection of 1 g/kg glucose. Fasting glucose levels (C), percentage hematocrit(D), serum glucose levels (E), and serum insulin levels (F ) were measured. All measurements were performed at the end of week 12.Euglycemic–hyperinsulinemic clamps were performed in DIO mice fasted overnight after 2 weeks of EPO treatment (n = 5–8/group). G: GIR.H: Whole-body glucose fluxes, GIR, endogenous glucose production, and glucose disposal. I and J: Tissue 2-deoxyglucose updatemeasured during clamp. Clamp plasma glucose levels were 209 + 39 and 157 + 13 mg/dL in saline- and EPO-treated mice, respectively.Clamp plasma insulin levels were 6.6 + 2.3 ng/mL in saline-treated mice and 5.8 + 0.8 ng/mL EPO-treated mice. Results are shown asmean6 SEM for n = 8 mice per group, representative of three independent experiments with similar results. *P< 0.05. BAT, brown adiposetissue; EGP, endogenous glucose production; Epi, epididymal; Ing, inguinal; Rd, glucose disposal.

diabetes.diabetesjournals.org Alnaeeli and Associates 2419

Page 6: Erythropoietin Signaling: A Novel Regulator of White ...diabetes.diabetesjournals.org/content/diabetes/63/7/2415.full.pdf · hormone indispensable for erythropoiesis, has biologi-cal

expressing cells in SVF (Fig. 3G), EPO-treated ani-mals exhibited reduced Mv infiltration associated withdecreased EPO-R expression in WAT (Fig. 3J), provid-ing a further link between WAT Mv and EPO/EPO-Rresponse.

EPO/EPO-R Signaling Induces Stat3 Phosphorylationin Mv and Inhibits Their Inflammatory Response InVitroTo investigate EPO/EPO-R signaling in WAT Mv and itsdirect effects on their inflammatory responses, Mvsorted from obese SVF were cultured in EPO-coatedplates for 15 min, fixed, and analyzed by phosphoflow.Although p-Stat5 and p-Stat6 were not detected in WTMv upon EPO treatment (Fig. 4A), EPO induced Stat3phosphorylation (Fig. 4B). These findings were furtherconfirmed by Western blotting (data not shown). EPO-Rabsence in ΔEpoR Mv prevented their response to EPOas shown by the absence of p-Stat3 (Fig. 4B, top right)compared with WT (Fig. 4B, top left). Moreover, treat-ment of WAT Mv with EPO (5 units/mL) for 24 h reducedproinflammatory genes expression of TNF-a and iNOS andmarkedly increased anti-inflammatory cytokine IL-10 ex-pression (Fig. 4C and D), collectively confirming a directresponse by WAT Mv to EPO/EPO-R signaling.

EPO Treatment Reverses Obesity-Induced Shift in MvPopulation SubtypesThe effect of EPO administration on Mv populationsubtypes in WAT was examined. Using flow cytometry,EPO decreased MGL-12 and CD11c+ while increasingMGL-1+ Mv (Fig. 5A and B). qRT-PCR confirmed phe-notypic Mv shift (Fig. 5C and D), showing that EPOreduced iNOS and Il-1b and increased Fizz-1, Ppar-g,and Arg-1. Overall, EPO increased M2-like Mv and re-duced M1-like Mv numbers, thus identifying and con-firming its role in the regulation of not only Mvinfiltration, but also local subtype polarization. Whetheror not the conversion of M1- to M2-like Mv, as recentlysuggested (46), contributes to the observed EPO-mediatedincrease in MGL-1+ Mv remains unknown. Since M1/M2nomenclature of Mv is based on in vitro studies, and maynot represent in vivo Mv subtypes (17), we use the des-ignation M1- and M2-like Mv to describe F4/80+MGL-12

and MGL-1+ cells, respectively.

Cellular Responses Drive EPO-Mediated Reversal ofMv Subtype ShiftM1 Mv infiltrating the WAT during DIO originate fromcirculating Ly6ChiCCR2+ inflammatory monocytes in aCCL2/CCR2-dependent manner (5,6,8). EPO lowers Ccl2

Figure 2—Continued

2420 Erythropoietin and Obesity-Induced Inflammation Diabetes Volume 63, July 2014

Page 7: Erythropoietin Signaling: A Novel Regulator of White ...diabetes.diabetesjournals.org/content/diabetes/63/7/2415.full.pdf · hormone indispensable for erythropoiesis, has biologi-cal

expression levels in WAT (Fig. 3C). We predicted EPO-mediated reduction of M1-like Mv subset (Fig. 5A and B)to be associated with fewer circulating Ly6ChiCCR2+ mono-cytes and reduced local WAT and serum CCL2 levels. Thiswas confirmed by flow cytometric analysis of whole blood

(Fig. 6A and B) and measurement of local WAT and serumCCL2 protein levels (Fig. 6C).

M2 Mv have been suggested to self-generate via pro-liferation in the presence of IL-4 in vivo (46,47). To studyhow the observed EPO-mediated expansion of M2-like

Figure 3—EPO-R expression profiles and the effects of EPO treatment on WAT inflammation and Mv infiltration. A: EPO-R expression inskeletal muscle, liver, and WAT of WT mice on normal chow (lean) was assessed using splenocytes and ΔEpoR WAT as positive andnegative controls, respectively. B: EPO-R expression in adipocytes and SVF was assessed in lean versus obese mice. Representative H&E-stained sections from perigonadal adipose tissue (C ) and immunofluorescent staining of perigonadal WAT sections for Mv (F4/80-red) andnuclei (DAPI-blue) (D) are shown; similar results are seen in 12 independent samples. E: Expression of inflammatory cytokine and chemo-kine genes in WAT from perigonadal fat was analyzed by qRT-PCR; expression levels are normalized to b-actin, and fold change inexpression are relative to negative control lean + saline. F: Serum TNF-a and IL-10 levels were determined for each group (n = 5 mice pergroup). G: Comparison of EPO-R levels in Mv (F4/80+) and non-Mv (F4/802) fractions, sorted by FACS, from stromal vascular cells of leanand obese WAT. Flow cytometry analysis of F4/80 expression in SVF cells of perigonadal WAT depicting total Mv percentage (H ) andnumber per gram of perigonadal WAT ( I ) are shown. J: EPO-R expression levels in SVF of lean and obese mice treated with and withoutEPO were determined by qRT-PCR. Expression levels were normalized to b-actin. Data are mean 6 SEM for n = 5 mice per group,representative of three independent experiments with similar results. *P < 0.05; **P < 0.01.

diabetes.diabetesjournals.org Alnaeeli and Associates 2421

Page 8: Erythropoietin Signaling: A Novel Regulator of White ...diabetes.diabetesjournals.org/content/diabetes/63/7/2415.full.pdf · hormone indispensable for erythropoiesis, has biologi-cal

Mv may occur (Fig. 5), quantitative comparisons of EPO-and saline-treated obese animals were performed. Ki-67staining and BrdU uptake showed that EPO promotesMGL-1+ Mv proliferation (Fig. 6D) in association withan increase in serum and WAT IL-4 (Fig. 6E). We con-firmed this in obese Stat62/2 and IL-42/2 mice, reportedpreviously to have defective M2-like Mv (48–50). EPOtreatment failed to expand M2-like Mv in these animals(Fig. 6F) unlike controls (Figs. 5B and 6F). These find-ings highlight a previously unrecognized role for EPO/EPO-R in the regulation of Mv polarization in vivo,

where IL-4/Stat6 axis is indispensable for EPO-mediatedM2-like Mv expansion.

Endogenous EPO/EPO-R Signaling Regulates MvInfiltration and Subtype ShiftTo investigate the role of endogenous EPO/EPO-R signal-ing during obesity-induced WAT inflammation, we usedDIO in ΔEpoR mice (39). The absence of EPO-R expres-sion in ΔEpoR WAT (adipocytes and SVF) and Mv wereconfirmed (Figs. 3B and 7A). ΔEpoR mice maintained onnormal chow become obese and glucose intolerant as they

Figure 3—Continued

2422 Erythropoietin and Obesity-Induced Inflammation Diabetes Volume 63, July 2014

Page 9: Erythropoietin Signaling: A Novel Regulator of White ...diabetes.diabetesjournals.org/content/diabetes/63/7/2415.full.pdf · hormone indispensable for erythropoiesis, has biologi-cal

age (36). Baseline measurements prior to the onset ofHFD feeding showed ΔEpoR mice (6–8 weeks old) tohave slightly higher body weight and fat mass comparedwith age-matched WT controls (Fig. 7B). However, after12 weeks of HFD feeding, there was no significant differ-ence in final body weight or fat mass between ΔEpoR andtheir age-matched WT controls (Fig. 7B).

Mv express the highest level of EPO-R mRNA amongimmune cell subsets profiled here (Fig. 7C) and are theonly EPO responsive population of immune cells in WTanimals (Supplementary Table 2). ΔEpoR animals withDIO showed higher circulating inflammatory monocyte

numbers (Fig. 7D), suggesting a possible direct role forendogenous EPO/EPO-R signaling in the regulation ofWAT inflammation independently of its effect on bodyweight and fat mass. ΔEpoR SVF cell chemokine andcytokine expression profiles showed elevated levels ofCcl1, Ccl2, Ccl12, Ccl17, Ccl19, Ccl22, Cxcl10, andCxcl11, with unchanged Ccl3, TNF-a, and IL-10 (Fig. 7E).Protein profiling confirmed the upregulation of these che-mokines, including CCL3 (data not shown). Despite un-changed TNF-a and IL-10 mRNA levels, we found higherTNF-a and similar IL-10 protein levels in ΔEpoR ani-mals (Fig. 7F).

Figure 4—EPO/EPO-R signaling induces Stat3 phosphorylation in Mv and inhibits their inflammatory response in vitro. WAT Mv, purifiedby FACS from obese mice SVF (after 12 weeks of HFD feeding), were cultured with saline or EPO (5 units/mL) for phosphoflow analysis.Histogram plots of Mv phosphoflow results for p-Stat5a/b (A) and p-Stat3 and p-Stat6 in WT (left two panels) and ΔEpoR (right two panels)(B). In some experiments, Mv sorted from SVF of obese WT mice were cultured with EPO (5 units/mL) for 24 h, after which their TNF-a andiNOS (C) and IL-10 (D) mRNA levels were quantified. Data represent observations from three independent experiments with similar resultsplotted as mean 6 SEM for n = 4 per group. *P < 0.05.

diabetes.diabetesjournals.org Alnaeeli and Associates 2423

Page 10: Erythropoietin Signaling: A Novel Regulator of White ...diabetes.diabetesjournals.org/content/diabetes/63/7/2415.full.pdf · hormone indispensable for erythropoiesis, has biologi-cal

CCL2 levels in ΔEpoR exceeded those in WT controls(Fig. 7F), where the difference in local WAT CCL2, but notserum, was significant (P # 0.05). Flow cytometry andhistology showed increased total Mv, denser inflammatoryinfiltrates and crown-like–structure formation, and moreMGL-12 Mv and fewer MGL-1+ Mv in WAT (Fig. 7G–I).The latter was confirmed via qRT-PCR (Fig. 7J). Despitereduced M2-like Mv numbers in WAT of ΔEpoR (Fig. 7I),serum and WAT IL-4 levels were not lower than WT con-trols (Fig. 7K). Based on Ki-67 staining the percentage ofproliferating MGL-1+ cells in ΔEpoR SVF was lower thantheir WT counterparts (data not shown).

Endogenous EPO/EPO-R Signaling Regulates Obesity-Induced Insulin Resistance and Glucose IntoleranceTo investigate the role of endogenous EPO/EPO-R signal-ing in regulating glucose metabolism, we compared WTwith ΔEpoR mice after 12 weeks of HFD feeding. ΔEpoR

mice showed higher insulin resistance, glucose intoler-ance, fasting blood glucose, and serum insulin (Fig. 8A–D).ΔEpoR mice maintained high insulin resistance after 2weeks of EPO administration (Fig. 8E and F), and therewas no change in the area under the curve and no signif-icant difference in glucose intolerance after EPO treatment(Fig. 8G). Not surprisingly, Mv infiltration and subtypecomposition in WAT of ΔEpoR mice remained similar afterEPO administration (Fig. 8H).

DISCUSSION

Herein, we characterized the role of EPO/EPO-R duringobesity-induced WAT inflammation and insulin resis-tance. Our observations reveal novel and unexpectedrole for EPO/EPO-R in regulating M1-like Mv infiltra-tion, M2-like Mv expansion in WAT, and inflammatorymonocyte recruitment (Figs. 3 and 5–7) prior to any body

Figure 5—EPO treatment regulates Mv subtype composition in WAT. SVF cells from perigonadal fat were used for flow cytometry andanalyses. Dot plots depict flow cytometry analysis of MGL-1+, MGL-12, and CD11c+ Mv subsets (A), and their numbers per gram of WAT areshown (B). Expression levels of iNOS and IL-1b (C) and Arg-1, Fizz-1, and Ppar-g (D) relative to b-actin were assessed. Results are shown asmean 6 SEM for n = 5 mice per group, representative of three independent experiments with similar results. *P < 0.05; **P < 0.01.

2424 Erythropoietin and Obesity-Induced Inflammation Diabetes Volume 63, July 2014

Page 11: Erythropoietin Signaling: A Novel Regulator of White ...diabetes.diabetesjournals.org/content/diabetes/63/7/2415.full.pdf · hormone indispensable for erythropoiesis, has biologi-cal

Figure 6—EPO treatment decreases circulating inflammatory monocytes and promotes WAT M2-like Mv expansion. Dot plots for flowcytometry analysis of circulating blood inflammatory monocytes Ly6ChiCCR2+ (A) and their numbers per milliliter of blood (B) are shown, aswell as CCL2 levels in serum and perigonadal WAT lysates (C). D: Flow cytometry results depict the percentage of proliferating MGL-12

versus MGL-1+ Mv in perigonadal SVF, based on Ki-67 staining (left) and BrdU uptake (right). E: Serum and WAT IL-4 levels are shown. F:Numbers of total Mv (bar) and MGL-1+ (top, black) and MGL-12 (bottom, gray) Mv subsets per gram of perigonadal WAT fromWT, Stat62/2,and IL-42/2 mice are shown. Results are presented as mean 6 SEM for n = 4–6 mice per group, representative of two or three independentexperiments with similar results. *P < 0.05; **P < 0.01.

diabetes.diabetesjournals.org Alnaeeli and Associates 2425

Page 12: Erythropoietin Signaling: A Novel Regulator of White ...diabetes.diabetesjournals.org/content/diabetes/63/7/2415.full.pdf · hormone indispensable for erythropoiesis, has biologi-cal

weight or fat mass loss (Fig. 1). We propose that the anti-inflammatory effects of EPO in WAT occur in associationwith its regulation of systemic glucose metabolism andthat it is not restricted to exogenous high-dose EPO treat-ment, as endogenous EPO is sufficient to regulate WATinflammation.

We and others have reported the reduction in bloodglucose with high-dose EPO treatment and transgenicoverexpression using mouse models of DIO and insulinresistance (35–38). The improved glucose metabolism isassociated with reduced body weight gain and fat massaccumulation, where EPO administration was initiated at

Figure 7—Endogenous EPO/EPO-R signaling regulates WAT Mv infiltration and subtype shift. WT C57BL/6 and age-matched ΔEpoR malemice with obesity induced by HFD feeding for 12 weeks were used. A: EPO-R expression levels were determined in SVF relative to b-actin.Shown are body weight and fat mass before and after DIO (B) and EPO-R expression analysis by qRT-PCR in different immune cell subsets(C) from spleens in which Mv were FACS-purified based on F4/80 expression and dendritic cells, B-cells, and T-cells that were purified bymagnetic activated cell sorting through positive selection of CD11c+ (dendritic cells), CD19+ (B-cells), and CD3+ (T-cells) cells. Shown arepercentage and numbers of circulating blood inflammatory monocytes (D); cytokine and chemokine gene expression profile of perigonadalSVF (E ); protein levels of TNF-a, IL-10, and CCL2 (F ); percentage and numbers of total Mv (G); and representative H&E-stained histologysections of perigonadal WAT (H ). MGL-1+, MGL-12, and CD11c+ Mv subset percentages (dot plots) and numbers per gram of perigonadalfat tissue ( I ), gene expression determined relative to b-actin (J ), and serum and WAT IL-4 levels (K) are shown. Results are mean 6 SEMfor n = 5 mice per group, representative of three independent experiments with similar results. *P < 0.05; **P < 0.01 WT vs. ΔEpoR. DC,dendritic cells.

2426 Erythropoietin and Obesity-Induced Inflammation Diabetes Volume 63, July 2014

Page 13: Erythropoietin Signaling: A Novel Regulator of White ...diabetes.diabetesjournals.org/content/diabetes/63/7/2415.full.pdf · hormone indispensable for erythropoiesis, has biologi-cal

the onset of HFD feeding and continued for 3 weeks orlonger (35–37). In stark contrast, herein, EPO adminis-tration was initiated after DIO, and its associated insulinresistance and glucose intolerance have already beenestablished and followed a 2-week duration regimen. Wereport improvements in glucose metabolism that precedeany detectable reduction in body weight or fat mass(Fig. 1), suggesting the EPO-mediated effects observedhere are independent from the role in body weight andfat mass regulation.

The shift to increased fat oxidation in muscle has beenshown to contribute to the observed normalization ofglucose sensitivity with EPO (38). These studies are con-sistent with our finding that EPO increased glucose up-take in skeletal muscle. Moreover, EPO was shown to

protect against diabetes through direct effects on pancre-atic b-cells in mouse models of types 1 and 2 diabetes notincluding DIO (36,37,40).

Herein, anti-inflammatory effect of EPO is identifiedand its signaling in Mv and effect on their activation andinfiltration are characterized. Indeed, it is known thatobesity-induced WAT inflammation is linked to increasedsystemic inflammation and insulin resistance (1–4). Wedemonstrate that EPO/EPO-R signaling in WAT Mv pro-motes Stat3 phosphorylation with clear anti-inflammatoryeffects (Fig. 4B and C), confirming a direct role in theregulation of Mv response. Interestingly, Stat3 phos-phorylation does not appear to be sufficient, in the ab-sence of Stat6, to mediate EPO/EPO-R signaling effect onM2-like Mv, as EPO treatment failed to expand this Mv

Figure 7—Continued

diabetes.diabetesjournals.org Alnaeeli and Associates 2427

Page 14: Erythropoietin Signaling: A Novel Regulator of White ...diabetes.diabetesjournals.org/content/diabetes/63/7/2415.full.pdf · hormone indispensable for erythropoiesis, has biologi-cal

subset in WAT of obese Stat62/2 mice (Fig. 6F), demon-strating a requirement for IL-4/Stat6 axis during EPO-mediated M2-like Mv expansion. In contrast, EPOtreatment markedly reduced total and M1-like WATMv populations in obese Stat62/2 mice and IL-42/2

mice (Fig. 6F), indicating that EPO/EPO-R/p-Stat3 axismay be sufficient to inhibit the proinflammatory response.Collectively, we conclude that both direct (Fig. 4B and C)and indirect via IL-4/Stat6 (Fig. 6F) responses of WAT Mvto EPO/EPO-R are involved in mediating the observed anti-inflammatory effects (Figs. 3 and 5–7).

Considering our observations that Mv are EPO re-sponsive, we propose that a direct contribution of theirEPO/EPO-R signaling plays a major role in the inhibitionof obesity-induced WAT inflammation. However, involve-ment of indirect EPO/EPO-R signaling in adipocytes on

obesity-induced WAT inflammation cannot be entirely ex-cluded, and its further investigation is required. SinceEPO/EPO-R signaling appears to regulate TNF-a levels(Figs. 3 and 7), the change in TNF-a may be linked toEPO effects on insulin resistance and glucose intolerance(Figs. 2 and 8). The exact molecular networks driving thislink, such as the role of JAK/STAT pathway and Akt ac-tivation downstream of EPO-R signaling, including thepossibility of insulin sensitivity regulation by directly af-fecting signaling downstream of insulin receptor, requirefurther studies.

Antiapoptotic and tissue-protective activities havebeen suggested to drive the anti-inflammatory effects ofEPO indirectly rather than via direct modulation ofimmune cell function/response (32). Evidence in sup-port of direct EPO effects on immune cells also exists

Figure 7—Continued

2428 Erythropoietin and Obesity-Induced Inflammation Diabetes Volume 63, July 2014

Page 15: Erythropoietin Signaling: A Novel Regulator of White ...diabetes.diabetesjournals.org/content/diabetes/63/7/2415.full.pdf · hormone indispensable for erythropoiesis, has biologi-cal

Figure 8—Endogenous EPO/EPO-R signaling and glucose metabolism during DIO. A: For ITT, glucose levels were measured after in-traperitoneal injection of 1 unit/kg insulin. B: For GTT, glucose levels were measured after intraperitoneal injection of 1 g/kg glucose. Fastingglucose (C ) and serum insulin (D) levels were determined. ΔEpoR mice with DIO, induced by 12 weeks of HFD feeding, were injectedsubcutaneously with saline or EPO (1,000 units/kg) every 48 h for the final 2 weeks of HFD feeding, and hematocrit (E ), ITT (F), GTT (G), andflow cytometry analysis of perigonadal fat SVF cells (H ) were determined. All measurements were performed at the end of week 12. Datapresented as mean 6 SEM for n = 8 mice per group, representative of three independent experiments with similar results. *P < 0.05.

diabetes.diabetesjournals.org Alnaeeli and Associates 2429

Page 16: Erythropoietin Signaling: A Novel Regulator of White ...diabetes.diabetesjournals.org/content/diabetes/63/7/2415.full.pdf · hormone indispensable for erythropoiesis, has biologi-cal

(33,34). To this end, we found Mv to display the high-est level of EPO-R expression among SVF cells ofWAT (Fig. 3E) and among other immune cell subsets(Fig. 7C). Moreover, the percentage of Mv appearedto be the only responder to EPO treatment amongSVF leukocytes (Supplementary Table 2), and EPO/EPO-R signaling in Mv was found to directly regulatetheir response, in agreement with the recently reportedanti-inflammatory role of EPO/EPO-R signaling duringbacterial infection and experimental colitis (33).

In summary, our studies demonstrate the involve-ment of EPO/EPO-R signaling in the reversal of insulinresistance and glucose intolerance; uncouple its role inobesity, body weight, and fat mass regulation from itseffect on inflammation; and collectively identify andcharacterize the anti-inflammatory effects of EPO. Wereveal a previously unrecognized role for EPO in theexpansion of M2-like Mv population via IL-4/Stat6axis and for Stat3 activation in the anti-inflammatoryeffects of EPO-R in WAT Mv. These findings highlighta novel EPO/EPO-R signaling-mediated regulation path-way of WAT inflammation in prediabetic condition,thus adding to its reported “anti-diabetic” effects shownto act via cytoprotection of pancreatic islet b-cells (40)or muscle fat oxidation (38) in experimental models ofdiabetes.

Acknowledgments. The authors thank William Jou at the National In-stitute of Diabetes and Digestive and Kidney Diseases Intramural Program MouseMetabolism Core Facility for his excellent technical assistance with metabolicstudies.Funding. This work was supported by the Intramural Research Program ofthe National Institute of Diabetes and Digestive and Kidney Diseases and theNational Institutes of Health.Duality of Interest. No potential conflicts of interest relevant to this articlewere reported.Author Contributions. M.A. designed experiments, conducted the stud-ies, analyzed the data, and wrote the manuscript. B.M.R., O.G., R.T., and T.C.contributed to data generation and analysis and reviewed and edited the man-uscript. C.T.N. contributed to experiment design and data discussion andreviewed and edited the manuscript. M.A. is the guarantor of this work and,as such, had full access to all the data in the study and takes responsibility forthe integrity of the data and the accuracy of the data analysis.

References1. Olefsky JM, Glass CK. Macrophages, inflammation, and insulin resistance.Annu Rev Physiol 2010;72:219–2462. Zeyda M, Stulnig TM. Obesity, inflammation, and insulin resistance—a mini-review. Gerontology 2009;55:379–3863. de Luca C, Olefsky JM. Inflammation and insulin resistance. FEBS Lett2008;582:97–1054. Lumeng CN, Saltiel AR. Inflammatory links between obesity and metabolicdisease. J Clin Invest 2011;121:2111–21175. Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch inadipose tissue macrophage polarization. J Clin Invest 2007;117:175–1846. Lumeng CN, Deyoung SM, Bodzin JL, Saltiel AR. Increased inflammatoryproperties of adipose tissue macrophages recruited during diet-induced obesity.Diabetes 2007;56:16–23

7. Wentworth JM, Naselli G, Brown WA, et al. Pro-inflammatory CD11c+CD206+adipose tissue macrophages are associated with insulin resistance in humanobesity. Diabetes 2010;59:1648–16568. Lumeng CN, DelProposto JB, Westcott DJ, Saltiel AR. Phenotypic switchingof adipose tissue macrophages with obesity is generated by spatiotemporaldifferences in macrophage subtypes. Diabetes 2008;57:3239–32469. Kanda H, Tateya S, Tamori Y, et al. MCP-1 contributes to macrophageinfiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity.J Clin Invest 2006;116:1494–150510. Kamei N, Tobe K, Suzuki R, et al. Overexpression of monocytechemoattractant protein-1 in adipose tissues causes macrophage recruitmentand insulin resistance. J Biol Chem 2006;281:26602–2661411. Arkan MC, Hevener AL, Greten FR, et al. IKK-beta links inflammation toobesity-induced insulin resistance. Nat Med 2005;11:191–19812. Odegaard JI, Ricardo-Gonzalez RR, Goforth MH, et al. Macrophage-specificPPARgamma controls alternative activation and improves insulin resistance.Nature 2007;447:1116–112013. Hevener AL, Olefsky JM, Reichart D, et al. Macrophage PPAR gamma isrequired for normal skeletal muscle and hepatic insulin sensitivity and fullantidiabetic effects of thiazolidinediones. J Clin Invest 2007;117:1658–166914. Mauer J, Chaurasia B, Plum L, et al. Myeloid cell-restricted insulin receptordeficiency protects against obesity-induced inflammation and systemic insulinresistance. PLoS Genet 2010;6:e100093815. Kiefer FW, Zeyda M, Gollinger K, et al. Neutralization of osteopontin inhibitsobesity-induced inflammation and insulin resistance. Diabetes 2010;59:935–94616. Féral CC, Neels JG, Kummer C, Slepak M, Olefsky JM, Ginsberg MH.Blockade of alpha4 integrin signaling ameliorates the metabolic consequences ofhigh-fat diet-induced obesity. Diabetes 2008;57:1842–185117. Westcott DJ, Delproposto JB, Geletka LM, et al. MGL1 promotes adiposetissue inflammation and insulin resistance by regulating 7/4hi monocytes inobesity. J Exp Med 2009;206:3143–315618. Nishimura S, Manabe I, Nagasaki M, et al. CD8+ effector T cells contributeto macrophage recruitment and adipose tissue inflammation in obesity. Nat Med2009;15:914–92019. Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM. Increasedadipose tissue expression of tumor necrosis factor-a in human obesity and in-sulin resistance. J Clin Invest 1995;95:2409–241520. Heilbronn LK, Campbell LV. Adipose tissue macrophages, low grade in-flammation and insulin resistance in human obesity. Curr Pharm Des 2008;14:1225–123021. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AWJr. Obesity is associated with macrophage accumulation in adipose tissue. J ClinInvest 2003;112:1796–180822. Ebert BL, Bunn HF. Regulation of the erythropoietin gene. Blood 1999;94:1864–187723. Semenza GL, Wang GL. A nuclear factor induced by hypoxia via de novoprotein synthesis binds to the human erythropoietin gene enhancer at a siterequired for transcriptional activation. Mol Cell Biol 1992;12:5447–545424. Semenza GL. Involvement of oxygen-sensing pathways in physiologic andpathologic erythropoiesis. Blood 2009;114:2015–201925. Sohmiya M, Kakiba T, Kato Y. Therapeutic use of continuous subcutaneousinfusion of recombinant human erythropoietin in malnourished predialysis anemicpatients with diabetic nephropathy. Eur J Endocrinol 1998;139:367–37026. Noguchi CT, Wang L, Rogers HM, Teng R, Jia Y. Survival and proliferativeroles of erythropoietin beyond the erythroid lineage. Expert Rev Mol Med 2008;10:e3627. Kertesz N, Wu J, Chen TH, Sucov HM, Wu H. The role of erythropoietin inregulating angiogenesis. Dev Biol 2004;276:101–11028. Wu H, Lee SH, Gao J, Liu X, Iruela-Arispe ML. Inactivation of erythropoietinleads to defects in cardiac morphogenesis. Development 1999;126:3597–360529. Yu X, Shacka JJ, Eells JB, et al. Erythropoietin receptor signalling is requiredfor normal brain development. Development 2002;129:505–516

2430 Erythropoietin and Obesity-Induced Inflammation Diabetes Volume 63, July 2014

Page 17: Erythropoietin Signaling: A Novel Regulator of White ...diabetes.diabetesjournals.org/content/diabetes/63/7/2415.full.pdf · hormone indispensable for erythropoiesis, has biologi-cal

30. Ueda K, Takano H, Niitsuma Y, et al. Sonic hedgehog is a critical mediator oferythropoietin-induced cardiac protection in mice. J Clin Invest 2010;120:2016–202931. Bernaudin M, Marti HH, Roussel S, et al. A potential role for erythropoietin infocal permanent cerebral ischemia in mice. J Cereb Blood Flow Metab 1999;19:643–65132. Villa P, Bigini P, Mennini T, et al. Erythropoietin selectively attenuates cy-tokine production and inflammation in cerebral ischemia by targeting neuronalapoptosis. J Exp Med 2003;198:971–97533. Nairz M, Schroll A, Moschen AR, et al. Erythropoietin contrastingly affectsbacterial infection and experimental colitis by inhibiting nuclear factor-kB-inducible immune pathways. Immunity 2011;34:61–7434. Yuan R, Maeda Y, Li W, Lu W, Cook S, Dowling P. Erythropoietin: a potentinducer of peripheral immuno/inflammatory modulation in autoimmune EAE.PLoS One 2008;3:e192435. Foskett A, Alnaeeli M, Wang L, Teng R, Noguchi CT. The effects of eryth-ropoietin dose titration during high-fat diet-induced obesity. J Biomed Biotechnol2011;2011:37378136. Teng R, Gavrilova O, Suzuki N, et al. Disrupted erythropoietin signallingpromotes obesity and alters hypothalamus proopiomelanocortin production. NatCommun 2011;2:52037. Katz O, Stuible M, Golishevski N, et al. Erythropoietin treatment leads toreduced blood glucose levels and body mass: insights from murine models.J Endocrinol 2010;205:87–9538. Hojman P, Brolin C, Gissel H, et al. Erythropoietin over-expression protectsagainst diet-induced obesity in mice through increased fat oxidation in muscles.PLoS ONE 2009;4:e589439. Suzuki N, Ohneda O, Takahashi S, et al. Erythroid-specific expression of theerythropoietin receptor rescued its null mutant mice from lethality. Blood 2002;100:2279–2288

40. Choi D, Schroer SA, Lu SY, et al. Erythropoietin protects against diabetesthrough direct effects on pancreatic beta cells. J Exp Med 2010;207:2831–284241. Wang R-H, Kim H-S, Xiao C, Xu X, Gavrilova O, Deng CX. Hepatic Sirt1deficiency in mice impairs mTorc2/Akt signaling and results in hyperglycemia,oxidative damage, and insulin resistance. J Clin Invest 2011;121:4477–449042. King IL, Dickendesher TL, Segal BM. Circulating Ly-6C+ myeloid precursorsmigrate to the CNS and play a pathogenic role during autoimmune demyelinatingdisease. Blood 2009;113:3190–319743. Krutzik PO, Nolan GP. Intracellular phospho-protein staining techniques forflow cytometry: monitoring single cell signaling events. Cytometry A 2003;55:61–7044. Kosteli A, Sugaru E, Haemmerle G, et al. Weight loss and lipolysis promotea dynamic immune response in murine adipose tissue. J Clin Invest 2010;120:3466–347945. Shoelson SE, Lee J, Goldfine AB. Inflammation and insulin resistance. J ClinInvest 2006;116:1793–180146. Jenkins SJ, Ruckerl D, Cook PC, et al. Local macrophage proliferation,rather than recruitment from the blood, is a signature of TH2 inflammation.Science 2011;332:1284–128847. Gordon S, Martinez FO. Alternative activation of macrophages: mechanismand functions. Immunity 2010;32:593–60448. Brombacher F, Arendse B, Peterson R, Hölscher A, Hölscher C. Analyzingclassical and alternative macrophage activation in macrophage/neutrophil-specific IL-4 receptor-a-deficient mice. Methods Mol Biol 2009;531:225–25249. Herbert DR, Hölscher C, Mohrs M, et al. Alternative macrophage activation isessential for survival during schistosomiasis and downmodulates T helper 1responses and immunopathology. Immunity 2004;20:623–63550. Takeda K, Tanaka T, Shi W, et al. Essential role of Stat6 in IL-4 signalling.Nature 1996;380:627–630

diabetes.diabetesjournals.org Alnaeeli and Associates 2431