14
Cécile Martinerie, 1,2 Marie Garcia, 1,2 Thi Thu Huong Do, 1,2 Bénédicte Antoine, 1,2 Marthe Moldes, 1,2 Guillaume Dorothee, 1 Chantal Kazazian, 1,2 Martine Auclair, 1,2 Marion Buyse, 1,2,3,4 Tatiana Ledent, 1 Pierre-Olivier Marchal, 1,2 Maria Fesatidou, 1,2 Adrien Beisseiche, 5 Haruhiko Koseki, 6 Shuichi Hiraoka, 7 Christos Evangelos Chadjichristos, 8 Bertrand Blondeau, 5 Raphael Georges Denis, 9 Serge Luquet, 9 and Bruno Fève 1,2,10 NOV/CCN3: A New Adipocytokine Involved in Obesity-Associated Insulin Resistance Diabetes 2016;65:25022515 | DOI: 10.2337/db15-0617 Identi cation of new adipokines that potentially link obesity to insulin resistance represents a major challenge. We recently showed that NOV/CCN3, a multifunctional matri- cellular protein, is synthesized and secreted by adipose tissue, with plasma levels highly correlated with BMI. NOV involvement in tissue repair, brotic and inammatory diseases, and cancer has been previously reported. How- ever, its role in energy homeostasis remains unknown. We investigated the metabolic phenotype of NOV 2/2 mice fed a standard or high-fat diet (HFD). Strikingly, the weight of NOV 2/2 mice was markedly lower than that of wild-type mice but only on an HFD. This was related to a signicant decrease in fat mass associated with an increased pro- portion of smaller adipocytes and to a higher expression of genes involved in energy expenditure. NOV 2/2 mice fed an HFD displayed improved glucose tolerance and insulin sensitivity. Interestingly, the absence of NOV was associ- ated with a change in macrophages prole (M1-like to M2- like), in a marked decrease in adipose tissue expression of several proinammatory cytokines and chemokines, and in enhanced insulin signaling. Conversely, NOV treatment of adipocytes increased chemokine expression. Altogether, these results show that NOV is a new adipocytokine that could be involved in obesity-associated insulin-resistance. More than 300 million people are currently obese world- wide, and this noninfectious epidemic affects both Western and emerging countries (13). In the absence of adequate care, obesity induces many comorbidities and reduces life expectancy (47). Thus, there is a crucial need to better understand the pathophysiology of obesity, insulin- resistance, and type 2 diabetes. In this context, the discovery of new adipocytokines involved in the control of energy balance is not only a challenge for understanding the mech- anisms responsible for the abnormal development of obesity or insulin resistance but also offers new opportunities to manage this disease and its complications. The NOV/CCN3 gene (nephroblastoma overexpressed gene) (8) is a founder of the CCN (Cyr61/CCN1, CTGF/CCN2, NOV/CCN3) family. The CCN genes encode matricial mul- tifunctional proteins that are involved in organogenesis (9). In addition, these proteins play key roles in inamma- tion (10), wound healing, brosis, and cancers (9). Several studies have also shown that NOV is involved in the ad- hesion, migration, proliferation, differentiation, and sur- vival of different cell types (1116). NOV also modulates the expression of inammatory molecules and their effects (1720). These different functions are mediated through 1 Sorbonne Universities, Pierre and Marie Curie University Paris 06, INSERM, Saint-Antoine Research Center, Saint-Antoine Hospital, Paris, France 2 Hospitalo-Universitary Institute, ICAN, Paris, France 3 Department of Pharmacy, Assistance Publique-Hôpitaux de Paris, Saint-Antoine Hospital, Paris, France 4 Paris-Sud University, EA 4123, Châtenay-Malabry, France 5 Sorbonne Universities, Pierre and Marie Curie University Paris 06, INSERM, Cordeliers Research Center, Paris, France 6 RIKEN Research Center for Allergy and Immunology (RCAI), RIKEN Yokohama Institute, Yokohama, Japan 7 Department of Biochemistry, Kobe Pharmaceutical University, Kobe, Japan 8 Sorbonne Universities, Pierre and Marie Curie University Paris 06, INSERM, Tenon Hospital, Paris, France 9 Sorbonne Paris City University, Paris Diderot University, BFA, CNRS, Paris, France 10 Department of Endocrinology, Paris, Assistance Publique-Hôpitaux de Paris, Saint-Antoine Hospital, Paris, France Corresponding author: Bruno Fève, [email protected]. Received 9 May 2015 and accepted 23 May 2016. This article contains Supplementary Data online at http://diabetes .diabetesjournals.org/lookup/suppl/doi:10.2337/db15-0617/-/DC1. © 2016 by the American Diabetes Association. Readers may use this article as long as the work is properly cited, the use is educational and not for prot, and the work is not altered. More information is available at http://diabetesjournals .org/site/license. 2502 Diabetes Volume 65, September 2016 METABOLISM

NOV/CCN3: A New Adipocytokine Involved in Obesity ... · (24). Taken together, these results and a recent report (25) showing that NOV is a direct target of transcription factor FOXO1

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: NOV/CCN3: A New Adipocytokine Involved in Obesity ... · (24). Taken together, these results and a recent report (25) showing that NOV is a direct target of transcription factor FOXO1

Cécile Martinerie,1,2 Marie Garcia,1,2 Thi Thu Huong Do,1,2 Bénédicte Antoine,1,2

Marthe Moldes,1,2 Guillaume Dorothee,1 Chantal Kazazian,1,2 Martine Auclair,1,2

Marion Buyse,1,2,3,4 Tatiana Ledent,1 Pierre-Olivier Marchal,1,2

Maria Fesatidou,1,2 Adrien Beisseiche,5 Haruhiko Koseki,6 Shuichi Hiraoka,7

Christos Evangelos Chadjichristos,8 Bertrand Blondeau,5 Raphael Georges Denis,9

Serge Luquet,9 and Bruno Fève1,2,10

NOV/CCN3: A New AdipocytokineInvolved in Obesity-Associated InsulinResistanceDiabetes 2016;65:2502–2515 | DOI: 10.2337/db15-0617

Identification of new adipokines that potentially link obesityto insulin resistance represents a major challenge. Werecently showed that NOV/CCN3, a multifunctional matri-cellular protein, is synthesized and secreted by adiposetissue, with plasma levels highly correlated with BMI. NOVinvolvement in tissue repair, fibrotic and inflammatorydiseases, and cancer has been previously reported. How-ever, its role in energy homeostasis remains unknown. Weinvestigated the metabolic phenotype of NOV2/2 mice feda standard or high-fat diet (HFD). Strikingly, the weight ofNOV2/2 mice was markedly lower than that of wild-typemice but only on an HFD. This was related to a significantdecrease in fat mass associated with an increased pro-portion of smaller adipocytes and to a higher expressionof genes involved in energy expenditure. NOV2/2 mice fedan HFD displayed improved glucose tolerance and insulinsensitivity. Interestingly, the absence of NOV was associ-ated with a change in macrophages profile (M1-like to M2-like), in a marked decrease in adipose tissue expression ofseveral proinflammatory cytokines and chemokines, and inenhanced insulin signaling. Conversely, NOV treatment ofadipocytes increased chemokine expression. Altogether,these results show that NOV is a new adipocytokine thatcould be involved in obesity-associated insulin-resistance.

More than 300 million people are currently obese world-wide, and this noninfectious epidemic affects both Westernand emerging countries (1–3). In the absence of adequatecare, obesity induces many comorbidities and reduceslife expectancy (4–7). Thus, there is a crucial need tobetter understand the pathophysiology of obesity, insulin-resistance, and type 2 diabetes. In this context, the discoveryof new adipocytokines involved in the control of energybalance is not only a challenge for understanding the mech-anisms responsible for the abnormal development of obesityor insulin resistance but also offers new opportunities tomanage this disease and its complications.

The NOV/CCN3 gene (nephroblastoma overexpressedgene) (8) is a founder of the CCN (Cyr61/CCN1, CTGF/CCN2,NOV/CCN3) family. The CCN genes encode matricial mul-tifunctional proteins that are involved in organogenesis(9). In addition, these proteins play key roles in inflamma-tion (10), wound healing, fibrosis, and cancers (9). Severalstudies have also shown that NOV is involved in the ad-hesion, migration, proliferation, differentiation, and sur-vival of different cell types (11–16). NOV also modulatesthe expression of inflammatory molecules and their effects(17–20). These different functions are mediated through

1Sorbonne Universities, Pierre and Marie Curie University Paris 06, INSERM,Saint-Antoine Research Center, Saint-Antoine Hospital, Paris, France2Hospitalo-Universitary Institute, ICAN, Paris, France3Department of Pharmacy, Assistance Publique-Hôpitaux de Paris, Saint-AntoineHospital, Paris, France4Paris-Sud University, EA 4123, Châtenay-Malabry, France5Sorbonne Universities, Pierre and Marie Curie University Paris 06, INSERM,Cordeliers Research Center, Paris, France6RIKEN Research Center for Allergy and Immunology (RCAI), RIKEN YokohamaInstitute, Yokohama, Japan7Department of Biochemistry, Kobe Pharmaceutical University, Kobe, Japan8Sorbonne Universities, Pierre and Marie Curie University Paris 06, INSERM,Tenon Hospital, Paris, France

9Sorbonne Paris City University, Paris Diderot University, BFA, CNRS, Paris, France10Department of Endocrinology, Paris, Assistance Publique-Hôpitaux de Paris,Saint-Antoine Hospital, Paris, France

Corresponding author: Bruno Fève, [email protected].

Received 9 May 2015 and accepted 23 May 2016.

This article contains Supplementary Data online at http://diabetes.diabetesjournals.org/lookup/suppl/doi:10.2337/db15-0617/-/DC1.

© 2016 by the American Diabetes Association. Readers may use this article aslong as the work is properly cited, the use is educational and not for profit, andthe work is not altered. More information is available at http://diabetesjournals.org/site/license.

2502 Diabetes Volume 65, September 2016

METABOLISM

Page 2: NOV/CCN3: A New Adipocytokine Involved in Obesity ... · (24). Taken together, these results and a recent report (25) showing that NOV is a direct target of transcription factor FOXO1

the interaction of NOV with specific integrins and, in somecases, by the Notch pathway (9).

Measurements of plasma NOV concentrations in a largecohort of patients screened for cardiometabolic diseases, inwhich 60% of the patients had a BMI above 30 kg/m2,revealed, for the first time, a strong relationship betweenplasma NOV concentrations and obesity (21). Plasma NOVlevels were also correlated with weight loss in patients whohad undergone bariatric surgery. Accordingly, we foundthat the NOV protein was synthesized and secreted byadipocytes and macrophages in adipose tissue from obesepatients and in human primary cultures (21). Moreover, wedemonstrated that the induction of NOV during adiposetissue expansion was not restricted to humans. Indeed, inmice fed a high-fat diet (HFD), plasma NOV levels and itsexpression in adipose tissue were also increased comparedwith mice fed a standard diet (SD) (21).

Even though a high BMI is often associated with im-paired glucose tolerance and type 2 diabetes, we did notfind any correlation between NOV and fasting glycemia.However, there was a positive correlation between plasmaNOV and HbA1c (21). Thus, it is conceivable that a rela-tionship could exist between NOV and carbohydrate me-tabolism. In this context, a possible link between NOVand insulin was suggested by Shimoyama et al. (22), whoreported that NOV expression was reduced in rat aortasafter streptozotocin injection and was increased by insulintreatment. Moreover, the nov gene is localized on chromo-some 8q24 (23), which is a susceptibility locus controllingb-cell function in linkage studies of patients with diabetes(24). Taken together, these results and a recent report (25)showing that NOV is a direct target of transcription factorFOXO1 and that NOV impairs insulin secretion in pancre-atic b-cells reinforce the possibility that this protein couldbe involved in energy homeostasis. However, no data havebeen reported to date on the function of NOV in adiposetissue or in whole-body metabolism. Thus, the main focusof this study was to identify whether targeted disruption ofthe nov gene modulates energy balance in mice fed a nor-mal or obesogenic diet.

RESEARCH DESIGN AND METHODS

MiceNOV2/2 mice were generated by Shimoyama et al. (22)using a targeting vector to delete the genomic regionencompassing exons 1, 2, and a part of 3 for completeelimination of the NOV transcription. Mice heterozygouslytransmitting the NOV-knockout genome were generated,and their descendants were backcrossed to C57Bl/6J forat least six generations and maintained as heterozy-gotes. C57Bl/6J heterozygous males were then mated with129sv/PAS females. Embryos derived from these crossingswere reimplanted in pseudopregnant females to get an F1on a mixed background (C57Bl/6J-SV129) with the “free ofspecific pathogenic microorganisms” status.

C57Bl/6-SV129 NOV2/2 mice and the wild-type (WT)controls were derived from the same heterozygous crossings.

Mice were kept on a 12-h light/12-h dark cycle with adlibitum access to food and tap water. Male mice (6 weeksold) were fed a SD (4% fat; Genestil 1326 Royaucourt,France) or an HFD (42% fat; TD.88137; Harlan Teklad,Gannat, France) for 16 weeks. Mice were bred accordingto the Guide for the Care and Use of Laboratory Animals(U.S. National Institutes of Health Publication No. 85-23, re-vised 1996). The animal facility was granted approval (B-75-12-01) given by the French Administration. All procedureswere approved by a local ethic committee (No. Ce5/2012/091).

Mice genotypes were performed as previously described(22). The genotypes were further checked by NOV mRNAlevels in epididymal white adipose tissue (eWAT) and sub-cutaneous WAT (scWAT), the plasma level of the proteinand its expression in heart where NOV is normally highlyexpressed (Supplementary Fig. 1).

Body CompositionBody weight (BW) was measured using a laboratory scale(Ohaus CS 200 Series; Sigma-Aldrich, l’Isle d’Abeau, France).Body mass composition was analyzed using an EchoMRI100 Whole Body Composition Analyzer (EchoMRI, Houston,TX) according to the manufacturer’s instructions (26).

Indirect Calorimetry MeasurementsFor measurement of energy expenditure, respiratory ex-change ratio (RER), and spontaneous locomotor activity,mice were placed in a LabMaster indirect calorimetrysystem (TSE Systems GmbH, Bad Homburg, Germany).

Metabolic Parameter ExplorationOral glucose tolerance tests (OGTTs) and insulin tolerancetests (ITTs) were performed after 14 and 15 weeks of thediet, respectively, on food-deprived (6 h and 4 h, respec-tively) nonanesthetized mice. For the OGTT, animalswere weighed and orally fed using gavage needles (Ref18060-020; Phymep, Paris, France) with a 1 g/kg BW of a20% glucose solution. Whole-tail vein blood (30 mL) wassampled at baseline and 15 min to measure plasma in-sulin. For the ITT, animals were weighed and intraperito-neally injected with human insulin (0.75 mU/kg). Forboth tests, glucose levels were obtained from whole-tail vein blood using an automatic Accu-Chek Performaglucometer (Roche, Meylan, France) at indicated times.For insulin-signaling experiments, mice were intraperitone-ally injected with 0.5 units of regular human insulin(Actrapid Penfill; Novo Nordisk, Paris, France). Tissueswere snap frozen in liquid nitrogen 10 min later.

Plasma MeasurementsTotal cholesterol, HDL and LDL cholesterol, triglycerides,and nonesterified fatty acids (NEFA) levels were de-termined by enzymatic colorimetric assays (SOBIODA,Montbonnot–Saint-Martin, France). Leptin, adiponectin,and NOV levels were measured using ELISA-specific kits(R&D Systems, Lille, France).

Pancreatic MeasurementsThe pancreatic and islet insulin content was extractedat 220°C in acidic ethanol (1.5% [vol/vol] HCl in 75%

diabetes.diabetesjournals.org Martinerie and Associates 2503

Page 3: NOV/CCN3: A New Adipocytokine Involved in Obesity ... · (24). Taken together, these results and a recent report (25) showing that NOV is a direct target of transcription factor FOXO1

[vol/vol] ethanol). Pancreatic islets from NOV2/2 and WTmice were isolated after collagenase digestion and culturedin RPMI-1640 containing 10% FBS at 37°C for 24 h. Batchesof 30 islets were incubated in Krebs buffer containing2.8 mmol/L glucose, 16.7 mmol/L glucose, or 50 mmol/LKCl for 1 h at 37°C. Plasma insulin, total pancreatic and isletinsulin content, and insulin secreted in response to glu-cose were measured using the Ultra Sensitive InsulinELISA kit (Crystal Chem, Inc., Downers Grove, IL).

Histomorphological ProceduresParaffin-embedded adipose sections were analyzed afterhematoxylin and eosin staining. Five fields were randomlyobserved at original magnification310 by an optical micro-scope (BX61 Olympus). Adipocyte quantification was per-formed using ImageJ software (http://rsbweb.nih.gov/ij/)on ;500–600 cells per mouse (4 animals per group andper genotype were analyzed).

Considering that adipocytes constituted 90% of theadipose tissue volume, the number of cells was estimatedby dividing the tissue mass by the mean volume of cells.The results are expressed in cells per gram of tissue.

Sections (3-mm thick) were stained with Sirius Red, andinterstitial fibrosis was assessed semiquantitatively byrandomly selecting 10 fields per section that coveredthe entire surface at original magnification 3200. Fibrosiswas then quantified using computer-based morphometricAnalysis software (Olympus) that allowed the formation ofa binary image in which the stained area could be automat-ically calculated as a percentage of the image area (27).

Isolation of Stromal Vascular Fraction and FlowCytometry AnalysiseWAT from WT and NOV2/2 mice was removed, minced,and digested with collagenase A (1 mg/mL in PBS;Sigma-Aldrich) at 37°C for 45 min. Homogenates werepassed through a 70-mm mesh, and the effluent wascentrifuged at 500g for 3 min. Cells from pellets definedas the stromal vascular fraction (SVF) were resuspendedin FACS buffer (PBS with 5% FBS). Extracellular stainingwas preceded by incubation with FcR-blocking antibody(2.4G2; BD Biosciences, Franklin Lakes, NJ) to avoidnonspecific staining. For lymphocytes cytometry analy-sis, cells were first stained with monoclonal antibodies tocell-surface markers purchased from BD Biosciences; fluo-rescein isothiocyanate–conjugated anti-CD3 (145-2C11),allophycocyanin-conjugated anti-CD4 (RM4-5), and peridinin-chlorophyll protein–conjugated anti-CD8 (53-6.7). Cellswere then fixed, permeabilized, and incubated with phyco-erythrin (PE)-conjugated anti-Foxp3 (FJK-16s, eBioscience)for intracellular staining according to the manufacturer’sspecifications. For staining analysis of macrophages, cellswere surface-stained with BV421-conjugated anti-F4/80(T45-2342), Alexa Fluor 647–conjugated anti-CD206 (19.2),and biotin-conjugated anti-CD11b (M1/70) revealed withPE-conjugated streptavidin all from BD Bioscience. Fluo-rescein isothiocyanate-conjugated anti-CD11c (N418) andanti–NOS2-PE cyanine 7 were from eBioscience. Stained

cells were analyzed using a Gallios flow cytometer(Beckman Coulter, Inc., Villepinte, France), and data wereprocessed using Kaluza software (Beckman Coulter).

Adipose Tissue and Liver Biochemical MeasurementseWAT from WT and NOV2/2 mice was removed anddigested as described above. Homogenates were centri-fuged, and isolated adipocytes were obtained after cen-trifugation. Glycerol secreted from isolated adipocyteswas used as an index of lipolysis and measured with theFree-Glycerol reagent (Sigma-Aldrich). Triacylglycerols wereextracted from adipocytes by the Dole method (28) andfrom liver by acetone and then tested with the triglycer-ides reagent (Sigma-Aldrich).

Primary Preadipocytes and 3T3-L1 CulturePrimary preadipocytes derived from SVF of scWAT oreWAT were plated on six-well culture dishes and culturedin DMEM (4.5 g/L D-glucose) with 10% FCS (Eurobio, LesUlis, France). Cell proliferation was assessed by BrdU in-corporation according to the manufacturer’s instructions(Cell Signaling, Danvers, MA). Adipocyte differentiation wasinitiated in confluent cultures of primary and 3T3-L1 pre-adipocytes with DMEM supplemented with 10% FCS and0.1 mmol/L 3-isobutyl-1-methylxanthine, 200 nmol/L dexa-methasone, and 100 nmol/L insulin (Sigma-Aldrich) for3 days. Adipocytes were further maintained for 6 daysin DMEM supplemented with 10% FCS and 100 nmol/Linsulin.

Primary Macrophages CultureBone marrow mononuclear (BMM) phagocytic precursorcells were isolated from femurs and tibiae of HFD-fed WTand NOV2/2 mice, as previously described (29). Theseprecursors were differentiated into adherent mature mac-rophages (BMM) for 6 days in noncoated Petri dishes inDMEM (4.5 g/L D-glucose) with 20% decomplementedFCS (Eurobio) containing 20 ng/mL macrophage colony-stimulating factor (PeproTech, Neuilly-sur-Seine, France).BMM were seeded at 2 3 106 cells/well in six-well platesand polarized toward a M1 phenotype with 10 ng/mLlipopolysaccharide (LPS; Sigma-Aldrich) or toward a M2phenotype with 10 ng/mL interleukin 4 (IL-4; PeproTech)for 24 h. Cytokine secretion into the cell culture mediumof BMM was subsequently analyzed using tumor necrosisfactor a (TNF-a) for M1 or IL-10 for M2 phenotype ELISAkit (PeproTech).

Peritoneal macrophages (PEM) were derived from HFD-fed WT and NOV2/2 mice after an intraperitoneal ad-ministration of PBS 13 (5 mL). The peritoneal wash wasrecovered from injected mice, and PEM were collected bycentrifugation at 600g at 4°C for 5 min. PEM, at a density of106 cells/well in a six-well plate, were polarized toward anM1 or an M2 phenotype as described for BMM. BMM orPEM were then processed as mentioned for FACS analysis.

Gene Expression AnalysisTotal RNA was extracted from mouse tissues, primarypreadipocytes, adipocyte cultures, and from 3T3-L1 cells

2504 NOV/CCN3 Is Involved in Insulin Resistance Diabetes Volume 65, September 2016

Page 4: NOV/CCN3: A New Adipocytokine Involved in Obesity ... · (24). Taken together, these results and a recent report (25) showing that NOV is a direct target of transcription factor FOXO1

using RNeasy Lipid Mini or RNeasy Mini Kits (Qiagen, LesUlis, France). Reverse transcription and real-time semi-quantitative PCR (qPCR) amplification on an ABI 7300 ap-paratus (Applied Biosystems) were performed as previouslydescribed (13). Specific primers (Proligo; Sigma-Aldrich)used for the amplification of target genes are presented inSupplementary Table 1. The comparative Ct method (30)was used to calculate gene expression values. The ribosomalS26 was used for normalization as a housekeeping gene.

Small Interfering RNA and TransfectionSmall interfering RNAs (siRNA) sequences predesigned byQiagen are listed in Supplementary Table 2. 3T3-L1 cellsplated at 9 3 103 cells/cm2 in DMEM containing 10% FCSwere transfected, 1 day later, with siRNA (120 pmol in 5 mL)using the RNAi Max reagent (Qiagen). Differentiation wasinitiated 72 h after transfection (day 0). On day 3 after theinduction of differentiation, total RNA extracted from thecells was subjected to real-time qPCR. For the study of insulinsignaling, 3T3-L1 cells were transfected on day 3 with siRNA(240 pmol in 10 mL); then, on day 5, the cultures were de-prived of serum and insulin and were further treated on day6 with insulin (1028 mol/L) for 8 min. RNA was obtainedfrom parallel cultures and underwent real-time qPCR.

Protein Analysis3T3-L1 adipocytes or mouse tissues were lysed, and equalprotein amounts (20 mg) were separated by SDS-PAGE.The conditioned medium corresponding to 106 3T3-L1 ad-ipocytes was used for ELISA tests or for NOV expression.

Antibodies and ChemicalsRecombinant human NOV protein was obtained as pre-viously described (15,17). For NOV protein detection, weused a homemade affinity-purified rabbit antibody (re-ferred as CT-m, anti-mouse NOV antibody) (31).

Rabbit polyclonal antibodies against total Akt, phos-phorylated (p)-Akt (P-Ser 473), total Erk, and p-Erk, werefrom Cell Signaling; uncoupling protein 1 (UCP-1), per-oxisome proliferator-activated receptor g (PPAR-g) coactiva-tor 1-a (PGC1-a), poly(ADP-ribose) polymerase 1 (Parp-1)were from Abcam (Paris, France), and mouse mono-clonal antibodies against b-actin, b-tubulin, and GAPDHwere from Sigma-Aldrich.

StatisticsThe results are expressed as the mean 6 SEM. Varianceequality was analyzed by an F test (Excel; Microsoft Corp.,Issy-Les-Moulineaux, France). Comparisons between groupsin the animal studies were carried out using a para-metric Student t test or by a nonparametric Mann-Whitney-Wilcoxon test (Minitab, Paris, France). A P valueof ,0.05 was considered statistically significant.

RESULTS

NOV2/2 Mice Are Protected Against HFD-InducedObesity, Glucose Intolerance, and Insulin ResistanceWe addressed the effect of NOV deficiency on weight gainin mice fed the SD or HFD for 16 weeks. No difference

was observed in mice fed the SD between the twogroups of mice (Fig. 1A and B). However, NOV2/2 micefed the HFD gained less weight than WT controls (Fig.1C). The BW gain of NOV2/2 mice was markedly lowercompared with WT mice (Fig. 1D). NOV2/2 mice fedthe HFD also displayed a decrease in fat and relativefat mass (Fig. 1E and F) and a slight increase in relativelean body mass compared with their respective con-trols; however, the absolute lean mass was unchanged(Fig. 1G and H), and there was no difference in mouse-tail length (96 6 0.83 mm for WT and 96.5 6 1.32 mmfor NOV2/2).

Indirect calorimetry was used to intensively analyzeenergy intake and energy expenditure of the mice. Nosignificant change in food intake (Fig. 1I) or in sponta-neous locomotor activity (Fig. 1K and SupplementaryFig. 2C) was observed. A slight increase in energy ex-penditure was detected in NOV2/2 mice but was limitedto the daylight period (Fig. 1J and Supplementary Fig.2B). The RER (VCO2-to-VO2 ratio) did not change be-tween NOV2/2 and WT mice (Fig. 1L and Supplemen-tary Fig. 2D).

To examine whether the marked differences in BWgain and adiposity between the two genotypes were asso-ciated with changes in carbohydrate metabolism, weevaluated glycemia and insulinemia under basal anddynamic conditions (Fig. 2). During the OGTT, HFD-fed(Fig. 2B) but not SD-fed (Fig. 2A) NOV2/2 animalsshowed improved glucose tolerance compared with WTmice. This correlated with a significant decrease in thearea under the curve for NOV2/2 mice compared withcontrols (Fig. 2B, upper panels). Of note, fasting glucoselevels were slightly lower in HFD-fed NOV2/2 mice com-pared with their WT controls. We then conducted an in-traperitoneal ITT (Fig. 2C and D), which demonstrated animproved insulin sensitivity in HFD-fed NOV2/2 micecompared with their WT littermates (Fig. 2D). The im-proved glucose tolerance observed in HFD-fed NOV2/2

mice was not accompanied by an increased insulinogenicindex after the oral glucose challenge compared with theWT controls (Fig. 2E). Because a previous study reporteddecreased insulin release subsequent to NOV overexpres-sion in a rat b-cell line (25), insulin secretion was testedin islets isolated from WT and NOV2/2 mice fed theHFD. Insulin secretion similarly increased in control andNOV2/2 islets when exposed to 16.7 mmol/L glucose or50 mmol/L KCl compared with 5.5 mmol/L glucose (Fig.2F). The pancreatic and islet insulin contents were alsosimilar between the two genotypes. Further pancreaticmorphometry analysis showed that b-cell fraction, b-cellmass, mean islet density, and mean islet size were similarin NOV2/2 and WT mice fed the HFD (SupplementaryFig. 3).

Collectively, these observations show that on an HFD,NOV deficiency decreased weight gain and improved glu-cose tolerance and insulin sensitivity, while insulin pro-duction remained unchanged.

diabetes.diabetesjournals.org Martinerie and Associates 2505

Page 5: NOV/CCN3: A New Adipocytokine Involved in Obesity ... · (24). Taken together, these results and a recent report (25) showing that NOV is a direct target of transcription factor FOXO1

HFD-Fed NOV2/2 Mice Display Less Adipose Tissue, aHigher Proportion of Small Adipocytes, and ImprovedLipid and Hepatic ParametersOn the SD, the organ weights–to–BW ratios were notsignificantly different between the two genotypes (Table1). However, on the HFD, a striking and similar decreaseof these ratios was observed in NOV2/2 mice comparedwith the WT controls for all the WAT depots. We alsoassessed the adipocyte size in hematoxylin and eosin–stained eWAT and scWAT tissue from WT and NOV2/2

mice fed the HFD. Of interest, we observed that NOVdeficiency influenced the adipocyte size distribution inboth fat tissues (Fig. 3). The mean adipocyte surface areain eWAT and scWAT was markedly reduced in NOV2/2

compared with WT mice (Fig. 3A and B). Moreover, x2

analysis on more than 1,500 independent determina-tions showed that in both eWAT and scWAT, NOV2/2

displayed a higher proportion of small adipocytes comparedwith those of the WT mice (P , 0.0001) (Fig. 3C and D).Adipocyte cellularity was also evaluated in eWAT and scWAT

with the assumption that adipocytes represent ;90% of adi-pose tissue volume (32). The results revealed no difference inthe number of adipocytes in scWAT betweenWT and NOV2/2

mice (3.10 6 0.41 and 3.1 6 0.46 3 106 cells/g of tissue,respectively). In contrast, the adipocyte number in eWATwas significantly increased in NOV2/2 mice (3.36 0.47 vs.2.1 6 0.32 3 106 cells/g of tissue, respectively; P , 0.05).

We also analyzed several plasma metabolic parameters,which are summarized in Table 1. On the SD, no differ-ences could be detected between the two genotypes. In WTmice, the HFD induced a significant increase in the con-centrations of triglycerides, cholesterol (C), HDL-C, NEFA,and leptin. In NOV2/2 mice, the HFD also provoked anincrease in cholesterol, triglycerides, HDL-C, and leptin,but not NEFA. However, compared with WT, the increasesin cholesterol, HDL-C, and leptin were significantly reducedin NOV2/2 mice. A trend (P , 0.1) toward a lower con-centration of LDL-C was also observed in these mice.

Basal or induced lipolysis was also investigated in adipo-cytes isolated from eWAT fat depots from HFD-fed mice.

Figure 1—BW of WT and NOV2/2 (KO) mice (n = 5–9 for each group) during a 16-week time course with the SD (A) or HFD (C ). BW gain inWT and NOV2/2 mice fed the SD (B) or HFD (D). Mean fat content (E), lean body mass content (G), relative fat content (g/BW) (F), and rela-tive lean body mass (g/BW) (H) of WT and NOV 2/2 mice fed the HFD for 14 weeks (n = 5 for each group). I: Food intake. Whole energyexpenditure was expressed as kcal/kg/h (J), spontaneous locomotor activity (beam break/h) (K ), and RER (VCO2-to-VO2) (L) of WTand NOV2/2 mice fed the HFD for 14 weeks (n = 5 for each group) by the mean of 4 days and 4 nights. Values are expressed as themeans 6 SEM. *P < 0.05 and **P < 0.01 for NOV2/2 vs. WT mice.

2506 NOV/CCN3 Is Involved in Insulin Resistance Diabetes Volume 65, September 2016

Page 6: NOV/CCN3: A New Adipocytokine Involved in Obesity ... · (24). Taken together, these results and a recent report (25) showing that NOV is a direct target of transcription factor FOXO1

As shown in Supplementary Fig. 4, basal and isoproterenol-stimulated lipolysis was stimulated in adipocytes fromNOV2/2 compared with those of WT animals.

The liver phenotype of WT and NOV2/2 mice fed theHFD was also characterized. Analysis of hepatic histology,illustrated in Supplementary Fig. 5A, revealed less steato-sis in NOV2/2 mice. Indeed, the hepatic triacylglycerol

content was reduced by approximately threefold (Supple-mentary Fig. 5B). This observation was consistent withdecreased mRNA levels of the fatty acid transporterCD36 and its main transactivator PPAR-g (SupplementaryFig. 5C). By contrast, no significant difference was de-tected for genes involved in fatty acid oxidation (PPAR-a,ACOX1), lipogenesis (SREBP-1c, ACC) or neoglucogenesis

Figure 2—Glucose tolerance, insulin sensitivity, and insulin secretion in WT and NOV2/2 (KO) mice. OGTT (A and B) and ITT (C and D) wereperformed after 14 and 15 weeks, respectively, for animals fed the SD (A and C) or HFD (B and D). The area under the curve (AUC) for OGTTmice (n = 10–14 per group) fasted for 6 h is presented in the upper panels of A and B. The results are expressed as the means 6 SEM.C and D: ITT mice (n = 8–11 per group) were fasted for 4 h. The results are expressed as the percentage of basal glycemia, with theAUC presented in the upper panels. E: The insulinogenic index was determined at 0 and 15 min after oral feeding with glucose and expressedas the ratio Ins15min-Ins0 (mU/L)–to–G15min-G0 (mmol/L). F: Insulin secretion assayed after in vitro incubation of islets in 5.5 mmol/L,16.7 mmol/L glucose, or 50 mmol/L KCl (n = 4–5 per group). *P < 0.05 and ***P < 0.001 for NOV2/2 vs. WT mice.

diabetes.diabetesjournals.org Martinerie and Associates 2507

Page 7: NOV/CCN3: A New Adipocytokine Involved in Obesity ... · (24). Taken together, these results and a recent report (25) showing that NOV is a direct target of transcription factor FOXO1

(PEPCK1, G6Pase). Interestingly, we did not detect anychange in several liver inflammation markers, includingchemokine (C-C motif) ligand 2 (CCL2), TNF-a, CD68,and F4/80 (Supplementary Fig. 5D).

In Vitro Preadipocyte Differentiation Is Increasedby NOV DeficiencyBecause a difference in adipogenesis could at least partlyexplain the reduced adipocyte size and adipose massfound in HFD-fed NOV2/2 mice, we first investigatedNOV expression in differentiating 3T3-L1 cells. As shownin Supplementary Fig. 6A, NOV mRNA and protein ex-pression was strongly induced during differentiation. Inagreement with this observation, there was an ;10-foldincrease in the NOV transcript level after adipocyte dif-ferentiation in primary cultures (Supplementary Fig. 6B).These findings suggested that NOV could be involved inadipogenesis. Thus, we then analyzed the expression ofthe three major transcription factors induced during adi-pogenesis in eWAT, perirenal WAT (prWAT), and scWAT.As shown in Fig. 4A–C, we did not find any significantdifferences in the expression of SREBP-1c, PPAR-g, andC/EBP-a between WT and NOV2/2 mice in the three fatdepots, but the pattern of these mRNAs did not excludethe possibility that NOV could modulate adipogenesis. Toexamine whether an alteration in adipocyte differentia-tion was an intrinsic property of NOV-deficient mice, weisolated SVFs from eWAT and scWAT and tested theirin vitro adipogenic capacity. Interestingly, we showed(Fig. 4D and E) that preadipocytes originating fromNOV2/2 mice had a greater adipocyte differentiation abil-ity than those from WT mice, as assessed by the increase

in SREBP-1c, PPAR-g, and C/EBP-a expression. In linewith this result, when NOV expression was suppressedin 3T3-L1 preadipocytes by transfection with an siRNAspecific for NOV, the expression of SREBP-1c, PPAR-g,and C/EBP-a was also increased compared with thesame cells transfected with a nonsilencing siRNA (Fig.4F). We did not observe any difference in the prolif-eration capacity of preadipocytes derived from WT orNOV2/2 mice (Supplementary Fig. 6C).

These results show that under HFD, the relative re-sistance to obesity and insulin resistance of NOV2/2

mice might be related to a higher proportion of smalladipocytes that likely exhibit better intrinsic adipogenicability in the absence of NOV.

Inflammation Is Reduced in Adipose Tissue FromNOV2/2 MiceNOV has been involved in the regulation of various in-flammatory molecules (10,20) that are known to impairinsulin signaling and promote insulin resistance in meta-bolic tissues. Consistent with enhanced insulin action andsensitivity (Fig. 2), we observed a specific pattern of al-tered inflammatory gene expression in different adiposetissues from HFD-fed NOV2/2 mice (Fig. 5). In eWAT andprWAT, transcript levels of TNF-a, CCL2, and F4/80 weredecreased (Fig. 5A and B), whereas the expression of thesegenes did not differ from WT in scWAT (Fig. 5C). We alsodetected a decrease in leptin levels in eWAT and scWATin NOV2/2 animals, consistent with their reduced leptinplasma levels (Table 1). Thus, in NOV2/2 mice fed theHFD, the reduced proinflammatory pattern of gene ex-pression essentially affects the visceral fat depots, which

Table 1—Organ weight and biological parameters in WT and NOV2/2 mice under SD and HFD

SD HFD

WT (n = 7) NOV2/2 (n = 10) WT (n = 9) NOV2/2 (n = 9)

Organ weight (mg)/BW (g)Liver 43.53 6 1.6 43.88 6 1.0 40.92 6 1.4 42.95 6 1.4Spleen 4.16 6 0.7 4.14 6 0.4 2.82 6 0.4 3.22 6 0.3Kidney 7.91 6 0.6 7.82 6 0.5 6.23 6 0.3 7.53 6 0.4*Pancreas 7.23 6 0.6 7.44 6 0.5 8.15 6 0.4 7.59 6 0.6prWAT 2.88 6 0.6 3.44 6 0.6 9.44 6 1.1‡‡‡ 4.84 6 0.9**eWAT 6.45 6 0.9 5.90 6 0.9 22.81 6 1.71‡‡‡ 13.19 6 1.4***‡‡‡scWAT 6.71 6 0.8 7.72 6 0.7 16.74 6 2.2‡‡ 9.13 6 1.7*BAT 4.1 6 0.7 3.83 6 0.6 6.56 6 0.7‡ 4.8 6 0.5

Biological parametersGlucose (mmol/L) 6.88 6 0.1 6.71 6 0.1 9.23 6 0.25‡‡‡ 8.35 6 0.3*‡‡‡Insulin (mUI/L) ND ND 16.04 6 2.82 15.21 6 1.29Cholesterol (g/L) 0.748 6 0.05 0.683 6 0.02 1.79 6 0.1‡‡‡ 1.51 6 0.1*‡‡‡Triglycerides (g/L) 0.481 6 0.03 0.472 6 0.03 2.65 6 0.1‡‡‡ 2.35 6 0.1‡‡‡HDL-C (g/L) 0.726 6 0.06 0.715 6 0.054 1.55 6 0.1‡‡‡ 1.29 6 0.1**‡‡‡LDL-C (g/L) ND ND 0.118 6 0.05 0.063 6 0.04NEFA (mmol/L) 168 6 16 165 6 31 259 6 30‡ 158 6 20**Leptin (ng/mL) 0.08 6 0.04 0.04 6 0.03 1.5 6 0.3‡‡‡ 0.66 6 0.2*‡‡Adiponectin (mg/mL) 2.11 6 0.2 1.93 6 0.3 2.42 6 0.2 2.25 6 0.2

All of these parameters were obtained at sacrifice after 16 weeks of the SD or HFD diet. Results are expressed as means 6 SEM. ND,not determined. *P , 0.05, **P , 0.01, and ***P , 0.001 for NOV2/2 vs. WT mice (within the same diet); ‡P , 0.05, ‡‡P , 0.01,and ‡‡‡P , 0.001 for HFD vs. SD mice (within the same genotype).

2508 NOV/CCN3 Is Involved in Insulin Resistance Diabetes Volume 65, September 2016

Page 8: NOV/CCN3: A New Adipocytokine Involved in Obesity ... · (24). Taken together, these results and a recent report (25) showing that NOV is a direct target of transcription factor FOXO1

are generally considered to be crucial in the control ofmetabolic status. Consistent with these data, we foundthat treatment of 3T3-L1 cells with NOV recombinantprotein led to a striking increase in CCL2 expression atboth the mRNA and protein level (Fig. 5D and E).

It is now largely recognized that rodent or humanobesity is associated with a low-grade inflammation ofadipose tissue and its infiltration by immune cells, includ-ing lymphocytes and macrophages, that shift from aM2-like anti-inflammatory to a M1-like proinflammatoryphenotype (33). In addition, these changes in macrophagephenotypes are likely involved in obesity-linked insulinresistance (34). Given the metabolic phenotype and theprofile of gene expression in deep fat depots of NOV2/2

animals, we further characterized the immune cell popu-lations present in eWAT of NOV2/2 and WT mice. Theabsence of NOV was associated with an increase in apopulation of macrophages expressing high levels of theM2-like marker CD206 (Fig. 5F) and with a trend to fewermacrophages displaying an apparent M1-like profile(CD11c+). No difference in the overall frequency of CD3+

lymphocytes population was detected between the twogenotypes (data not shown). Although no differencewas observed in the frequency of CD8+T cells, the per-centages of CD4+ lymphocytes among infiltrating T cellswere strongly decreased in NOV2/2 compared with WT(Fig. 5G, left panel). This was related to significantly re-duced frequency of conventional CD4+Foxp3– T cells, with

no difference in percentages of CD4+Foxp3+ regulatoryT cells (Fig. 5G, right panel). Interestingly, a large pro-portion of infiltrating CD3+ lymphocytes were identifiedCD4–CD8– (double-negative) T cells, the frequency of whichwas significantly increased in NOV2/2 compared with WTmice (Fig. 5G, left panel).

We further analyzed whether NOV deletion can affectM1 versus M2 polarization in myeloid/macrophage cells.Macrophages were derived from BMM (or PEM) of WTand NOV2/2 HFD-fed mice. NOV expression was report-ed in human macrophages (21); however, its expression inmacrophages from HFD-fed WT mice has not been inves-tigated so far. In our experimental conditions, the levelsof NOV mRNA in BMM or PEM derived from HFD-fedWT mice were below the threshold of detection (data notshown). Nevertheless, because a transient expression ofNOV during macrophage maturation or differentiationcould affect their susceptibility to proinflammatory mole-cules, we subjected WT and NOV2/2 BMM or PEM culturesto polarization to a M1-like or M2-like phenotype with LPSand IL-4, respectively (32). Compared with untreated con-trol cultures, the mRNA levels of M1-like proinflammatorymarkers, such as TNF-a and CCL2, were significantly in-duced in BMM cultures treated with LPS, whereas IL-4markedly enhanced the M2-like marker CD206 (Supple-mentary Fig 7A). We also observed in our experimentalconditions, as previously described (35), that the mRNAlevels of the M2-like marker IL-10 was considerably

Figure 3—Adipocyte size and cellularity in WT and NOV2/2 (KO) mice fed the HFD for 16 weeks. A and B: Representative hematoxylin andeosin–stained sections of eWAT and scWAT. Adipocyte size distribution determined from eWAT (C) and scWAT (D) histomorphometry andimage analysis; insets show the mean adipocyte surface 6 SEM. ***P < 0.001. A x2 test was performed for each histogram; ***P < 0.0001for NOV2/2 vs. WT animals.

diabetes.diabetesjournals.org Martinerie and Associates 2509

Page 9: NOV/CCN3: A New Adipocytokine Involved in Obesity ... · (24). Taken together, these results and a recent report (25) showing that NOV is a direct target of transcription factor FOXO1

induced by LPS. Analysis of TNF-a and IL-10 proteins con-firmed the mRNA results (Supplementary Fig. 7B).

Consistent with these results, we also found that amongthe CD11b+F4/80+ population of BMM treated with LPSthere was a significant increase of macrophages expressingthe M1-like marker inducible nitric oxide synthase 2,whereas IL-4 markedly enhanced the percentage of cellsexpressing the M2-like marker CD206 (SupplementaryFig. 7C). Overall, mRNA, secretion, or FACS analyses de-tected no significant difference between WT and NOV2/2

BMM under these different treatments (SupplementaryFig. 7A–C). Similar results were also obtained with PEM(data not shown). Thus, whereas NOV2/2 were protectedagainst inflammation in adipose tissue, it does not seem tobe due to the absence of NOV in macrophages.

Because limited inflammation and reduced fibrosis werepreviously described in NOV2/2 mice in the unilateral ure-teral obstruction experimental model of nephropathy (36)and because fibrosis in adipose tissue is also linked toinsulin-resistance (37), we also studied the effect of NOVdeletion on fibrosis of the adipose tissue. Analysis of theinterstitial collagen accumulation, assessed by Sirius Red col-oration, revealed a trend for decreased fibrosis in eWAT ofNOV2/2 animals and no significant difference between WTand NOV2/2 mice in the scWAT (Supplementary Fig. 8).

Expression of Genes Involved in Energy ExpenditureIs Increased by NOV Deficiency in Adipose TissueA slight increase in energy expenditure was observed inNOV 2/2 mice (Fig. 1J). Because an increased turnover of

adipose cells in these mice could be energetically moreexpensive, we first analyzed markers of apoptosis in adi-pose tissue. Western blots did not reveal any significantdifferences in the Parp-1 cleavage in scWAT of both ge-notypes on the HFD, whereas Parp-1 cleavage in eWATwas even decreased in NOV2/2 animals (SupplementaryFig. 9). Thus, these results could not account for the lowerBW gain of NOV2/2 mice.

We next analyzed the expression of genes in the brownadipose tissue (BAT) and WAT known to be involved inenergy expenditure. In HFD-fed NOV2/2 mice, the ex-pression of UCP-1 was increased in BAT and eWAT atboth mRNA and protein levels (Fig. 6). The expressionof PGC1-a, a major coregulator that cooperates withPPAR-g to transactivate the UCP-1 gene promoter (38),was also significantly increased in the BAT of these mice(Fig. 6).

In Vitro and In Vivo NOV Deficiency Increases InsulinSensitivityTo further delineate the molecular mechanisms involvedin the improved insulin sensitivity in NOV2/2 mice, weanalyzed the effect of NOV deficiency on insulin signaling.We found that in 3T3-L1 adipocytes transfected with twodifferent siRNAs specific for NOV, the decrease in NOVexpression was accompanied by a marked increase ininsulin-stimulated p-Akt and p-Erk compared with thesame cells transfected with a nonsilencing siRNA as a con-trol (sc) (Fig. 7A–C). In vivo, we also observed that an acuteexposure to insulin resulted in a significant increase in

Figure 4—Expression of genes involved in adipocyte differentiation in eWAT (A), prWAT (B), and scWAT (C ) in WT and NOV2/2 (KO) micefed the HFD for 16 weeks (n = 6–8 per group). SVF derived from eWAT (D) and scWAT (E ) from mice fed the HFD for 16 weeks were culturedand underwent adipocyte differentiation for 7 days. F: Adipocyte differentiation was induced for 3 days in 3T3-L1 cells transfected withcontrol nonsilencing siRNA (SC) or a specific siRNA against NOV (Si NOV). In the inset, the inhibition of NOV expression was evaluated onday 3 after adipocyte differentiation. Data represent the mean values of four measurements and correspond to a representative experimentperformed at least three times with similar results. **P < 0.01 and ***P < 0.001 for NOV2/2 or siNOV conditions vs. control.

2510 NOV/CCN3 Is Involved in Insulin Resistance Diabetes Volume 65, September 2016

Page 10: NOV/CCN3: A New Adipocytokine Involved in Obesity ... · (24). Taken together, these results and a recent report (25) showing that NOV is a direct target of transcription factor FOXO1

p-Akt in eWAT of NOV2/2 compared with WT mice (Fig.7D and E) but not in scWAT (data not shown).

DISCUSSION

In this study, we report for the first time that NOVdeficiency protects mice fed an HFD against obesity, likelythrough an increase in energy expenditure. We also report

that in NOV2/2 mice on an HFD, the inflammation of theeWAT and prWAT was reduced compared with WT mice.Taken together, these differences between WT and NOV2/2

mice likely account for the improved glucose tolerance,insulin sensitivity, and metabolic profile of NOV2/2 mice.

The increased population of small adipocytes in eWATcould be due to the better proliferation or the better

Figure 5—Expression of inflammatory genes and analysis of infiltrating immune cell populations in eWAT (A), prWAT(B), and scWAT (C) ofWT and NOV2/2 (KO) mice fed the HFD for 16 weeks. After 24 h of serum starvation, 3T3-L1 adipocytes were incubated with vehicle (Ctr) orNOV (10 mg/mL) for 24 h, and the level of CCL2 expression was evaluated in cells by real-time qPCR (D) or in the conditioned media byELISA (E ). Data represent the mean values 6 SEM (n = 4–9 experiments). *P < 0.05, **P < 0.01, and ***P < 0.001 for KO mice or NOV-treated cells vs. control. F and G: Flow cytometry analysis of macrophages and lymphocytes isolated from eWAT of WT and NOV2/2 mice.F: Percentages of CD11c+ (left panel), or CD206+high (right panel) cells among CD11b+F4/80+ macrophages. G: Percentages of totalCD8+CD4+ or CD4–CD8– double negative (DN) T cells (left and middle panels) and CD4+Foxp3+ regulatory T cells or CD4+Foxp3– con-ventional T cells (right panel) among stromal CD3+ lymphocytes. Data represent mean values 6 SEM (n = 4 per group). *P < 0.05.

diabetes.diabetesjournals.org Martinerie and Associates 2511

Page 11: NOV/CCN3: A New Adipocytokine Involved in Obesity ... · (24). Taken together, these results and a recent report (25) showing that NOV is a direct target of transcription factor FOXO1

differentiation ability of the resident preadipocytes. Ourdata are in favor of the second hypothesis because we didnot observe any difference in the proliferation rate ofprimary preadipocytes derived from WT or NOV2/2 mice.In contrast, the absence of NOV had a positive effect onthe differentiation ability of these cells. Moreover, differ-ent data have reported that NOV could play an effectiverole in the control of cell differentiation because it caninhibit myoblast and osteoblast differentiation (13,39,40).It is also crucial for primitive hematopoietic progenitor cellactivity (14). However, although the improved ability todifferentiate in NOV2/2 preadipocytes was observed inboth eWAT and scWAT, the cellularity was only increasedin eWAT. This observation is in accordance with recentdata (41) showing that in mice fed an HFD for over1 month, adipogenesis is preferentially initiated in eWATcompared with scWAT. This potentially improved abilityfor preadipocyte differentiation, in which NOV is disruptedor inhibited, is apparently contradictory with the decreasedfat mass in NOV2/2 mice and with the increase in NOVexpression in vitro during the differentiation process. Ourresults suggest that the absence of NOV favors the recruit-ment of smaller adipocytes that are more sensitive to in-sulin. However, a longer follow-up of WT and NOV2/2

mice would be required to examine whether the recruit-ment of small adipocytes maintains the limited expansion

of fat mass in NOV2/2 mice or whether this is overcomeover time by increased cellularity.

Taking into account the whole body, this fat mass lossoccurs while there is no decrease in food intake, andpreliminary analysis of lipids in feces did not reveal anysignificant difference in gut absorption of lipids betweenNOV2/2 and control mice (Supplementary Fig. 10). How-ever, there is a slight but significant increased energyexpenditure during the daylight period. Remarkably, thisenhanced energy expenditure is associated with an inductionin BAT and eWAT in the expression of UCP1 and PGC1-a,two key effectors of the thermogenic pathway. This couldrepresent a major phenomenon to protect NOV2/2 micefrom HFD-induced obesity and insulin resistance. However,the molecular mechanisms by which the absence of NOVleads to UCP1 induction remain unknown. For instance, theincreased lipolytic activity of NOV2/2 adipocytes could alsopromote substrate availability for thermogenesis.

In this study, we report that the effect of the absenceof NOV is only detectable when mice are challenged withan HFD. Indeed, two reports describing the phenotype ofNOV2/2 mice revealed that they are viable and appar-ently normal and fertile (22,40). However, after a lesionof the femoral artery, the null mice presented an enhance-ment of neointimal thickening compared with WT miceduring vascular repair (22). Another example showed that

Figure 6—Expression of genes and proteins involved in energy expenditure in BAT (A) and eWAT (B) of WT and NOV2/2 (KO) mice fed theHFD for 16 weeks (n = 6–8 per group). C and D: Representative Western blots of BAT and eWAT. E and F: Bar graphs show fold change inUCP1 and PGC1-a in KO vs. WT mice. The results are expressed as the means 6 SEM. *P < 0.05 and ***P < 0.001 for NOV2/2 vs. WTmice.

2512 NOV/CCN3 Is Involved in Insulin Resistance Diabetes Volume 65, September 2016

Page 12: NOV/CCN3: A New Adipocytokine Involved in Obesity ... · (24). Taken together, these results and a recent report (25) showing that NOV is a direct target of transcription factor FOXO1

after an injury to the femur, bone regeneration in NOV2/2

mice was accelerated compared with WT mice (40).More recently, we also found that during the progressionof obstructive nephropathy, NOV2/2 mice displayed lim-ited inflammation and renal interstitial fibrosis comparedwith WT mice (36).

An HFD is known to induce low-grade inflammation inadipose tissue (42) and is associated with the increased ex-pression of NOV mRNA in adipose tissue and NOV plasmalevels (21). Under these specific nutritional conditions, theabsence of NOV not only limits adipose tissue expansion butis also associated with improved glucose tolerance. This isprimarily related to better insulin sensitivity, as supportedby ITTs and by the normalization of plasma NEFA levels inNOV2/2 mice fed the HFD, suggesting the restoration ofthe antilipolytic effect of insulin. These results are strength-ened by in vitro and in vivo experiments showing that NOVdeficiency in adipocytes promotes insulin signaling.

Our results also strongly suggest that NOV could be akey player in adipose tissue inflammation and that this inturn could promote insulin resistance. Recent studieshave shown that CD4+ and CD8+ T cells play a major rolein promoting adipose tissue inflammation by secretingcytokines and recruiting macrophages during HFD (43).NOV deficiency led to a strong decrease in the frequencyof infiltrating conventional CD4+ T cells, whereas no dif-ference was observed for CD8+ or CD4+ regulatory T cells.

Of note, NOV2/2 mice displayed a significantly in-creased subpopulation of CD3+ T lymphocytes that were

CD4–CD8–. These particular T cells that lack the expres-sion of both CD4 and CD8 T-cell coreceptors have beenstudied in mice and humans for their contribution to peri-pheral tolerance and disease prevention (44). Our cur-rent findings are consistent with these observations. Thereduced TNF-a and F4/80 expression in two visceral WAT(eWAT and prWAT) depots from HFD-fed NOV2/2

mice reflects the decreased inflammation in these fatdepots and is in agreement with the involvement ofthese tissues in insulin sensitivity (45–47). Interestingly,a decrease in TNF-a and F4/80 expression was not de-tected in scWAT, which is generally considered to havean accessory role in the insulin resistance phenotype.FACS analysis of eWAT suggests that under HFD, theabsence of NOV was associated with a switched overbal-ance from M1-like to M2-like macrophage population.However, our in vitro analysis revealed that the absenceof NOV did not influence the polarization of macro-phages toward a M1 or M2 phenotype. This stronglysuggests that the switch from M1-like to M2-like mac-rophage population does not result from an autocrineaction of NOV on macrophages but rather from an in-direct role of NOV on the expression of proinflammatorymolecules by adipocytes and possibly also by other cellspresent in the adipose tissue. Given the documentedprotective role of such M2-like populations againstobesity-linked insulin resistance, this suggests that thesechanges may also contribute to the improved insulinsensitivity and glucose tolerance of NOV2/2 mice.

Figure 7—Insulin signaling in siNOV-transfected 3T3-L1 adipocytes and in eWAT from WT and NOV2/2 (KO) mice fed the HFD. A:Representative Western blots of 3T3-L1 adipocytes transfected with a control nonsilencing siRNA (sc) or with two different specific siRNAsagainst NOV (si1 and si4) from day 3 to 6 after adipocyte differentiation. B: Bar graphs show fold change in p-Akt/Akt and p-Erk/Erk in si1-and si4- vs. sc-transfected cells. Values are the means 6 SEM of three experiments. C: NOV gene expression as determined in parallelcultures. *P < 0.05 and ***P < 0.001 for si1- or si4-treated vs. sc-treated cells. D: Representative Western blots of eWAT from WT and KOmice after or not a 10-min exposure to insulin (0.5 units). E: Bar graphs show fold change in p-Akt/Akt. The results are expressed as themeans 6 SEM. ###P < 0.001 for NOV2/2 vs. WT mice; ***P < 0.001 for insulin-treated vs. untreated animals.

diabetes.diabetesjournals.org Martinerie and Associates 2513

Page 13: NOV/CCN3: A New Adipocytokine Involved in Obesity ... · (24). Taken together, these results and a recent report (25) showing that NOV is a direct target of transcription factor FOXO1

Our in vitro and in vivo experiments revealed thatCCL2 expression is modulated by NOV. This regulationmay be considered to be pivotal in the development ofinsulin resistance because targeted deletions in mice forthe CCL2 and/or CCR2 genes result in reduced macro-phage content, decreased inflammation in fat, and pro-tection from HFD-induced insulin resistance; conversely,mice overexpressing CCL2 in adipose tissue have in-creased local macrophage infiltration along with insulinresistance (47–49). The specific macrophage markerF4/80 expression is reduced in NOV2/2 mice WAT. Thissuggests that the absence of NOV could decrease CCL2with a subsequent reduction in macrophage number withinthe adipose tissue. However, the regulation of CCL2 byNOV could also have an effect on insulin sensitivitythrough additional pathways because it has also beenshown that an increase in the plasma levels of CCL2 issufficient to induce insulin resistance independent ofmacrophage infiltration into adipose tissue (50). Ac-cordingly, CCL2, through its angiogenic effect on endo-thelial cells, could also contribute to the expansion andremodeling of adipose tissue (51).

During the OGTT, we did not observe a higher plasmainsulin concentration in NOV2/2 mice compared with WTcontrols. Insulin release in isolated islets was also compa-rable between the two genotypes, and the total content ofinsulin in the whole pancreas or in isolated islets wassimilar in NOV2/2 and WT mice. These results are appar-ently discordant with those reported by Paradis et al. (25).Nevertheless, the experimental conditions used by thatstudy and ours substantially differ because they used arat cell line overexpressing NOV. Taken together, our re-sults suggest that the increase in insulin sensitivity is themajor mechanism by which glucose tolerance is improvedin NOV2/2 mice.

In summary, the current study demonstrates that NOVparticipates in the development of obesity-induced insu-lin resistance. These results indicate that the use of coun-teracting agents against NOV represents a potential strategyfor new therapies against obesity and its metaboliccomorbidities.

Acknowledgments. The authors acknowledge L. Dinard from the animalfacilities platform (PHEA Saint-Antoine) for her technical support, A. Munier fromthe cytometry platform (UMS, Lumic 030) for her help in FACS analysis,R. Morichon from the image platform (UMS, Lumic 030) for his help in micro-scopy analysis, and S. Dumont and F. Merabetene for their contribution inadipose tissue histomorphological analysis (UMS, Lumic 030). The authors alsoacknowledge the technical platform Functional and Physiological Exploration(FPE) of the Unit “Biologie Fonctionnelle et Adaptative” (Sorbonne Paris CityUniversity, Paris Diderot University, BFA, CNRS, Paris, France) for metabolicand behavioral analysis.Funding. This work was financed by INSERM, Pierre et Marie Curie University,and by a grant from the French Society of Diabetes. T.T.H.D. received financialsupport from the French Society of Endocrinology and the French Embassy inVietnam, and P.-O.M. received financial support from ANRT (National Associationof Research and Technology).

Duality of Interest. No potential conflicts of interest relevant to this articlewere reported.Author Contributions. C.M. and B.F. designed the research. M.G., T.T.H.D.,B.A., M.M., G.D., C.K., M.A., M.B., T.L., P.-O.M., M.F., A.B., B.B., and R.G.D.performed the research. H.K., S.H., and C.E.C. contributed new reagents/analytictools. C.M., B.A., M.M., G.D., B.B., R.G.D., S.L., and B.F. analyzed data. C.M. andB.F. wrote the paper. B.F. is the guarantor of this work and, as such, had fullaccess to all the data in the study and takes responsibility for the integrity of thedata and the accuracy of the data analysis.

References1. DeMaria EJ. Bariatric surgery for morbid obesity. N Engl J Med 2007;356:2176–21832. Yoon KH, Lee JH, Kim JW, et al. Epidemic obesity and type 2 diabetes inAsia. Lancet 2006;368:1681–16883. He J, Klag MJ, Whelton PK, Zhoa Y, Weng X. Short- and long-term prog-nosis after acute myocardial infarction in Chinese men and women. Am J Epi-demiol 1994;139:693–7034. Haslam DW, James WP. Obesity. Lancet 2005;366:1197–12095. Renehan AG, Tyson M, Egger M, Heller RF, Zwahlen M. Body-mass indexand incidence of cancer: a systematic review and meta-analysis of prospectiveobservational studies. Lancet 2008;371:569–5786. Renehan A, Smith U, Kirkman MS. Linking diabetes and cancer: a con-sensus on complexity. Lancet 2010;375:2201–22027. Romero-Corral A, Montori VM, Somers VK, et al. Association of bodyweightwith total mortality and with cardiovascular events in coronary artery disease:a systematic review of cohort studies. Lancet 2006;368:666–6788. Joliot V, Martinerie C, Dambrine G, et al. Proviral rearrangements andoverexpression of a new cellular gene (nov) in myeloblastosis-associated virustype 1-induced nephroblastomas. Mol Cell Biol 1992;12:10–219. Chen CC, Lau LF. Functions and mechanisms of action of CCN matricellularproteins. Int J Biochem Cell Biol 2009;41:771–78310. Kular L, Pakradouni J, Kitabgi P, Laurent M, Martinerie C. The CCN family: anew class of inflammation modulators? Biochimie 2011;93:377–38811. Laurent M, Martinerie C, Thibout H, et al. NOVH increases MMP3 expressionand cell migration in glioblastoma cells via a PDGFR-alpha-dependent mecha-nism. FASEB J 2003;17:1919–192112. Lin CG, Chen CC, Leu SJ, Grzeszkiewicz TM, Lau LF. Integrin-dependentfunctions of the angiogenic inducer NOV (CCN3): implication in wound healing.J Biol Chem 2005;280:8229–823713. Calhabeu F, Lafont J, Le Dreau G, et al. NOV/CCN3 impairs muscle cellcommitment and differentiation. Exp Cell Res 2006;312:1876–188914. Gupta R, Hong D, Iborra F, Sarno S, Enver T. NOV (CCN3) functions as a reg-ulator of human hematopoietic stem or progenitor cells. Science 2007;316:590–59315. Doghman M, Arhatte M, Thibout H, et al. Nephroblastoma overexpressed/cysteine-rich protein 61/connective tissue growth factor/nephroblastoma over-expressed gene-3 (NOV/CCN3), a selective adrenocortical cell proapoptotic factor,is down-regulated in childhood adrenocortical tumors. J Clin Endocrinol Metab2007;92:3253–326016. McCallum L, Lu W, Price S, Lazar N, Perbal B, Irvine AE. CCN3: a key growthregulator in chronic myeloid leukaemia. J Cell Commun Signal 2009;3:115–12417. Lafont J, Jacques C, Le Dreau G, et al. New target genes for NOV/CCN3 inchondrocytes: TGF-beta2 and type X collagen. J Bone Miner Res 2005;20:2213–222318. Riser BL, Najmabadi F, Perbal B, et al. CCN3 (NOV) is a negative regulator ofCCN2 (CTGF) and a novel endogenous inhibitor of the fibrotic pathway in anin vitro model of renal disease. Am J Pathol 2009;174:1725–173419. Chen CC, Young JL, Monzon RI, Chen N, Todorovi�c V, Lau LF. Cytotoxicity ofTNFalpha is regulated by integrin-mediated matrix signaling. EMBO J 2007;26:1257–126720. Le Dréau G, Kular L, Nicot AB, et al. NOV/CCN3 upregulates CCL2 andCXCL1 expression in astrocytes through beta1 and beta5 integrins. Glia 2010;58:1510–1521

2514 NOV/CCN3 Is Involved in Insulin Resistance Diabetes Volume 65, September 2016

Page 14: NOV/CCN3: A New Adipocytokine Involved in Obesity ... · (24). Taken together, these results and a recent report (25) showing that NOV is a direct target of transcription factor FOXO1

21. Pakradouni J, Le Goff W, Calmel C, et al. Plasma NOV/CCN3 levels areclosely associated with obesity in patients with metabolic disorders. PLoS One2013;8:e6678822. Shimoyama T, Hiraoka S, Takemoto M, et al. CCN3 inhibits neointimalhyperplasia through modulation of smooth muscle cell growth and migration.Arterioscler Thromb Vasc Biol 2010;30:675–68223. Martinerie C, Viegas-Pequignot E, Guénard I, et al. Physical mapping ofhuman loci homologous to the chicken nov proto-oncogene. Oncogene 1992;7:2529–253424. An P, Freedman BI, Rich SS, et al. Quantitative trait loci on chromosome8q24 for pancreatic beta-cell function and 7q11 for insulin sensitivity in obesenondiabetic white and black families: evidence from genome-wide linkage scansin the NHLBI Hypertension Genetic Epidemiology Network (HyperGEN) study.Diabetes 2006;55:551–55825. Paradis R, Lazar N, Antinozzi P, Perbal B, Buteau J. Nov/Ccn3, a noveltranscriptional target of FoxO1, impairs pancreatic b-cell function. PLoS One2013;8:e6495726. Taicher GZ, Tinsley FC, Reiderman A, Heiman ML. Quantitative magneticresonance (QMR) method for bone and whole-body-composition analysis. AnalBioanal Chem 2003;377:990–100227. Abed A, Toubas J, Kavvadas P, et al. Targeting connexin 43 protects againstthe progression of experimental chronic kidney disease in mice. Kidney Int 2014;86:768–77928. Dole VP, Meinertz H. Microdetermination of long-chain fatty acids in plasmaand tissues. J Biol Chem 1960;235:2595–259929. Stanley ER. Murine bone marrow-derived macrophages. Methods Mol Biol1997;75:301–30430. Pfaffl MW. A new mathematical model for relative quantification in real-timeRT-PCR. Nucleic Acids Res 2001;29:e4531. Kular L, Rivat C, Lelongt B, et al. NOV/CCN3 attenuates inflammatory painthrough regulation of matrix metalloproteinases-2 and -9. J Neuroinflammation2012;9:3632. Eto H, Suga H, Matsumoto D, et al. Characterization of structure and cellularcomponents of aspirated and excised adipose tissue. Plast Reconstr Surg 2009;124:1087–109733. Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch inadipose tissue macrophage polarization. J Clin Invest 2007;117:175–18434. Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Annu RevImmunol 2011;29:415–44535. Saraiva M, O’Garra A. The regulation of IL-10 production by immune cells.Nat Rev Immunol 2010;10:170–18136. Marchal PO, Kavvadas P, Abed A, et al. Reduced NOV/CCN3 expressionlimits inflammation and interstitial renal fibrosis after obstructive nephropathy inmice. PLoS One 2015;10:e0137876

37. Sun K, Tordjman J, Clément K, Scherer PE. Fibrosis and adipose tissuedysfunction. Cell Metab 2013;18:470–47738. Puigserver P, Wu Z, Park CW, Graves R, Wright M, Spiegelman BM. A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell1998;92:829–83939. Sakamoto K, Yamaguchi S, Ando R, et al. The nephroblastoma overex-pressed gene (NOV/ccn3) protein associates with Notch1 extracellular domainand inhibits myoblast differentiation via Notch signaling pathway. J Biol Chem2002;277:29399–2940540. Matsushita Y, Sakamoto K, Tamamura Y, et al. CCN3 protein participatesin bone regeneration as an inhibitory factor. J Biol Chem 2013;288:19973–1998541. Wang QA, Tao C, Gupta RK, Scherer PE. Tracking adipogenesis during whiteadipose tissue development, expansion and regeneration. Nat Med 2013;19:1338–134442. Chandalia M, Abate N. Metabolic complications of obesity: inflated or in-flamed? J Diabetes Complications 2007;21:128–13643. Huh JY, Park YJ, Ham M, Kim JB. Crosstalk between adipocytes and im-mune cells in adipose tissue inflammation and metabolic dysregulation in obe-sity. Mol Cells 2014;37:365–37144. Hillhouse EE, Lesage S. A comprehensive review of the phenotype andfunction of antigen-specific immunoregulatory double negative T cells. J Auto-immun 2013;40:58–6545. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AWJr. Obesity is associated with macrophage accumulation in adipose tissue. J ClinInvest 2003;112:1796–180846. Xu H, Barnes GT, Yang Q, et al. Chronic inflammation in fat plays a crucialrole in the development of obesity-related insulin resistance. J Clin Invest 2003;112:1821–183047. Weisberg SP, Hunter D, Huber R, et al. CCR2 modulates inflammatory andmetabolic effects of high-fat feeding. J Clin Invest 2006;116:115–12448. 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;116:1494–150549. Kamei N, Tobe K, Suzuki R, et al. Overexpression of monocyte chemo-attractant protein-1 in adipose tissues causes macrophage recruitment and in-sulin resistance. J Biol Chem 2006;281:26602–2661450. Tateya S, Tamori Y, Kawaguchi T, Kanda H, Kasuga M. An increase in thecirculating concentration of monocyte chemoattractant protein-1 elicits systemicinsulin resistance irrespective of adipose tissue inflammation in mice. Endocri-nology 2010;151:971–97951. Salcedo R, Ponce ML, Young HA, et al. Human endothelial cells expressCCR2 and respond to MCP-1: direct role of MCP-1 in angiogenesis and tumorprogression. Blood 2000;96:34–40

diabetes.diabetesjournals.org Martinerie and Associates 2515