6
Selective Autoantibody Production against CCL3 Is Associated with Human Type 1 Diabetes Mellitus and Serves As a Novel Biomarker for Its Diagnosis 1 Naim Shehadeh, ‡§ Shirly Pollack, § Gizi Wildbaum,* § Yaniv Zohar,* § Itay Shafat, § Reem Makhoul, Essam Daod, Fahed Hakim, Rina Perlman, and Nathan Karin 2 * †§ We have recently demonstrated that patients suffering from chronic autoimmune diseases develop an autoantibody response against key mediators that participate in the initiation and progression of these diseases. In this paper, we show that patients with type 1 diabetes mellitus (T1DM), but not those suffering from several other inflammatory autoimmune diseases, display a selective autoantibody titer to a single CC chemokine named CCL3. From the diagnostic point we show that this response could be used as a biomarker for diagnosis of T1DM, a disease that is currently diagnosed by autoantibodies to competitive anti-insulin Abs, islet cell Abs, and glutamic acid decarboxylase Abs. We show that our currently suggested biomarker is more reliable than each of the above alone, including diagnosis of T1DM at its preclinical stage, and could therefore be used as a novel way for diagnosis of T1DM. These Abs were found to be neutralizing Abs. It is possible, though hard to prove, that these Abs participate in the natural regulation of the human disease. Hence, it has previously been shown by others that selective neutralization of CCL3 suppresses T1DM in NOD mice. Theses results together with ours suggest CCL3 as a preferential target for therapy of T1DM. The Journal of Immunology, 2009, 182: 8104 – 8109. W e have recently shown that patients suffering from chronic autoimmune diseases develop an autoanti- body response against key mediators that participate in the initiation and regulation of these diseases. Thus, patients suffering from rheumatoid arthritis (RA), 3 but not osteoarthritis, develop a significant autoantibody titer against TNF- (1). Neu- tralizing Abs to TNF- suppress RA but not osteoarthritis (2). In humans, it is hard to prove that these Abs participate in the natural regulation of RA. Nevertheless, we showed that their elimination ag- gravated the manifestation of the experimentally induced disease in rodents (1). Thus, it is possible that in an attempt to restrain autoim- munity, the immune system can selectively promote autoantibody production in a selective manner. Using animal models of the disease, we found out that this response is turned on at the preclinical stage of the disease to ameliorate its pathological consequences, and is later regulated by the dynamics of the disease (3). Type 1 diabetes mellitus (T1DM) is the result of organ-specific autoimmune destruction of the insulin-secreting -cells in the pan- creatic islets of Langerhans (4). It has become evident that T1DM is a multifactorial disease mediated by T cells in which CD4 and CD8 T cells and macrophages are required for -cell destruction (5). Although the critical events that trigger the autoreactive pro- cess in T1DM are not clear, destruction of insulin-producing -cells appears to be mediated by the activation of autoreactive T cells that recognize several islet -cells Ags, including competitive anti-insulin Abs (CIAA), islet cell Abs (ICA), glutamic acid de- carboxylase (GAD) 65 and 67 isotypes Abs, heat shock protein 60, and some uncharacterized -cells Ags (6 – 8). The current diagno- sis of T1DM is based on measuring autoantibodies to GAD, ICA, ICA 512 (IA-2) and insulin (9 –17). Chemokines are small (8 –14 kDa), structurally related pro- teins that regulate cell trafficking through interactions with a subset of seven transmembrane, G protein-coupled receptors (18 –21). As such they are thought to be key mediators of inflammation, includ- ing inflammatory autoimmune diseases, and therefore valid targets for therapy. Based on the position of key cysteines, chemokine are divided to four subgroups: C, CC, CXC, and CX 3 C. Of these sub- groups, various CC chemokines, CXCL8 and CXCR3 ligands, mostly CXCL10, were strongly associated with inflammatory au- toimmunity, including T1DM, in experimental models (22–28). It is difficult to directly extrapolate these observations to patients with T1DM. In an attempt to indirectly identify key chemokines that could be associated with the dynamics of human T1DM, we have explored our recent target discovery strategy (1) from human RA to human T1DM. In this paper, we show that of the various chemokines that have been associated with inflammatory autoim- munity, patients with T1DM develop a selective autoantibody re- sponse to a single CC chemokines (CCL3), emphasizing its po- tential role as a major target not only for experimental models of disease but for therapy as well (25). The current study focuses on its potential use as a novel and effective diagnostic tool. Materials and Methods Study population The basic study population was comprised of 221 children. The study groups were divided into 87 new onset diabetic patients aged 2–18 years, who were admitted to the pediatric ward of Meyer Children’s Hospital *Department of Immunology, Rappaport Family Institute for Research in the Med- ical Sciences, and Rambam Medical Center, § Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel Received for publication October 6, 2008. Accepted for publication April 10, 2009. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. 1 The study was supported by the Israel Science Foundation and by a research grant of Micro-Medic (Israel). 2 Address correspondence and reprint requests to Dr. Nathan Karin, Bruce Rappaport Faculty of Medicine, Technion, P.O.B. 9697, Haifa 31096, Israel. E-mail address: [email protected] 3 Abbreviations used in this paper: RA, rheumatoid arthritis; CF, cystic fibrosis; CIAA, competitive anti-insulin Ab; GAD, glutamic acid decarboxylase; ICA, islet cell Ab; JRA, juvenile RA; ROC, receiver operating characteristic; T1DM, type 1 diabetes mellitus. Copyright © 2009 by The American Association of Immunologists, Inc. 0022-1767/09/$2.00 The Journal of Immunology www.jimmunol.org/cgi/doi/10.4049/jimmunol.0803348

Selective Autoantibody Production against CCL3 Is Associated with Human Type 1 Diabetes Mellitus and Serves As a Novel Biomarker for Its Diagnosis

Embed Size (px)

Citation preview

Selective Autoantibody Production against CCL3 Is Associatedwith Human Type 1 Diabetes Mellitus and Serves As a NovelBiomarker for Its Diagnosis1

Naim Shehadeh,‡§ Shirly Pollack,§ Gizi Wildbaum,*§ Yaniv Zohar,*§ Itay Shafat,§

Reem Makhoul,‡ Essam Daod,‡ Fahed Hakim,‡ Rina Perlman,‡ and Nathan Karin2*†§

We have recently demonstrated that patients suffering from chronic autoimmune diseases develop an autoantibody response against keymediators that participate in the initiation and progression of these diseases. In this paper, we show that patients with type 1 diabetesmellitus (T1DM), but not those suffering from several other inflammatory autoimmune diseases, display a selective autoantibody titerto a single CC chemokine named CCL3. From the diagnostic point we show that this response could be used as a biomarker for diagnosisof T1DM, a disease that is currently diagnosed by autoantibodies to competitive anti-insulin Abs, islet cell Abs, and glutamic aciddecarboxylase Abs. We show that our currently suggested biomarker is more reliable than each of the above alone, including diagnosisof T1DM at its preclinical stage, and could therefore be used as a novel way for diagnosis of T1DM. These Abs were found to beneutralizing Abs. It is possible, though hard to prove, that these Abs participate in the natural regulation of the human disease. Hence,it has previously been shown by others that selective neutralization of CCL3 suppresses T1DM in NOD mice. Theses results togetherwith ours suggest CCL3 as a preferential target for therapy of T1DM. The Journal of Immunology, 2009, 182: 8104–8109.

W e have recently shown that patients suffering fromchronic autoimmune diseases develop an autoanti-body response against key mediators that participate

in the initiation and regulation of these diseases. Thus, patientssuffering from rheumatoid arthritis (RA),3 but not osteoarthritis,develop a significant autoantibody titer against TNF-� (1). Neu-tralizing Abs to TNF-� suppress RA but not osteoarthritis (2). Inhumans, it is hard to prove that these Abs participate in the naturalregulation of RA. Nevertheless, we showed that their elimination ag-gravated the manifestation of the experimentally induced disease inrodents (1). Thus, it is possible that in an attempt to restrain autoim-munity, the immune system can selectively promote autoantibodyproduction in a selective manner. Using animal models of the disease,we found out that this response is turned on at the preclinical stage ofthe disease to ameliorate its pathological consequences, and is laterregulated by the dynamics of the disease (3).

Type 1 diabetes mellitus (T1DM) is the result of organ-specificautoimmune destruction of the insulin-secreting �-cells in the pan-creatic islets of Langerhans (4). It has become evident that T1DMis a multifactorial disease mediated by T cells in which CD4� and

CD8� T cells and macrophages are required for �-cell destruction(5). Although the critical events that trigger the autoreactive pro-cess in T1DM are not clear, destruction of insulin-producing�-cells appears to be mediated by the activation of autoreactive Tcells that recognize several islet �-cells Ags, including competitiveanti-insulin Abs (CIAA), islet cell Abs (ICA), glutamic acid de-carboxylase (GAD) 65 and 67 isotypes Abs, heat shock protein 60,and some uncharacterized �-cells Ags (6–8). The current diagno-sis of T1DM is based on measuring autoantibodies to GAD, ICA,ICA 512 (IA-2) and insulin (9–17).

Chemokines are small (�8–14 kDa), structurally related pro-teins that regulate cell trafficking through interactions with a subsetof seven transmembrane, G protein-coupled receptors (18–21). Assuch they are thought to be key mediators of inflammation, includ-ing inflammatory autoimmune diseases, and therefore valid targetsfor therapy. Based on the position of key cysteines, chemokine aredivided to four subgroups: C, CC, CXC, and CX3C. Of these sub-groups, various CC chemokines, CXCL8 and CXCR3 ligands,mostly CXCL10, were strongly associated with inflammatory au-toimmunity, including T1DM, in experimental models (22–28). Itis difficult to directly extrapolate these observations to patientswith T1DM. In an attempt to indirectly identify key chemokinesthat could be associated with the dynamics of human T1DM, wehave explored our recent target discovery strategy (1) from humanRA to human T1DM. In this paper, we show that of the variouschemokines that have been associated with inflammatory autoim-munity, patients with T1DM develop a selective autoantibody re-sponse to a single CC chemokines (CCL3), emphasizing its po-tential role as a major target not only for experimental models ofdisease but for therapy as well (25). The current study focuses onits potential use as a novel and effective diagnostic tool.

Materials and MethodsStudy population

The basic study population was comprised of 221 children. The studygroups were divided into 87 new onset diabetic patients aged 2–�18 years,who were admitted to the pediatric ward of Meyer Children’s Hospital

*Department of Immunology, †Rappaport Family Institute for Research in the Med-ical Sciences, and ‡Rambam Medical Center, §Bruce Rappaport Faculty of Medicine,Technion, Haifa, Israel

Received for publication October 6, 2008. Accepted for publication April 10, 2009.

The costs of publication of this article were defrayed in part by the payment of pagecharges. This article must therefore be hereby marked advertisement in accordancewith 18 U.S.C. Section 1734 solely to indicate this fact.1 The study was supported by the Israel Science Foundation and by a research grantof Micro-Medic (Israel).2 Address correspondence and reprint requests to Dr. Nathan Karin, Bruce RappaportFaculty of Medicine, Technion, P.O.B. 9697, Haifa 31096, Israel. E-mail address:[email protected] Abbreviations used in this paper: RA, rheumatoid arthritis; CF, cystic fibrosis;CIAA, competitive anti-insulin Ab; GAD, glutamic acid decarboxylase; ICA, isletcell Ab; JRA, juvenile RA; ROC, receiver operating characteristic; T1DM, type 1diabetes mellitus.

Copyright © 2009 by The American Association of Immunologists, Inc. 0022-1767/09/$2.00

The Journal of Immunology

www.jimmunol.org/cgi/doi/10.4049/jimmunol.0803348

(Haifa, Israel) between January 2004 and December 2007, 54 age-matchedhealthy subjects, 34 long-standing diabetic children (�5 years duration)who were visiting the diabetic pediatric clinic at Meyer Children’s Hos-pital, 20 subjects who were in prediabetic state (first-degree relatives of aT1DM patient and positive for at least one of the known autoantibodies;GAD, ICA, or CIAA), and 14 children with juvenile rheumatoid arthritis(JRA) who were being followed up by our rheumatologic pediatric clinic.All patients were diagnosed according to the following criteria: age at onset�16 years, arthritis in one or more joints, duration of disease 6 wk orlonger, exclusive of other forms of juvenile arthritis. An additional groupincluded 12 long-term patients with RA (seven women and five men, meanage 54, and range 26–73), disease duration: range 2–28 years, mean 13.8years.

All subjects diagnosed as RA patients were rheumatoid factor positive(seropositive), accompanied by erosive RA, and fulfilled all of the Amer-ican College of Radiology’s criteria for RA.

RA and JRA patients were on various nonsteroidal anti-inflammatorydrugs, low steroid therapy (maximum dose 10 mg/day), and had differentdisease-modifying drugs like Methotrexate, Plaquenil, Imuran, Solganal(gold injections), and anti TNF �.

Twenty cystic fibrosis (CF) patients (nine females) also participated inthis study; these patients are being followed up at the CF Center and thepediatric pulmonary unit within the Meyers Children’s Hospital. Ten pa-tients are pancreatic insufficient and the other ten are pancreatic sufficient.Their average age is 16.75 years (range 1–37 years) and all patients areon traditional CF medications, none of these patients require systemicsteroid treatment.

All studies were conducted according our protocol, which was approvedby our local Helsinki committee at the Rambam Medical Center. All sub-jects provided informed consent under this protocol.

The clinical service laboratory of endocrinology at Rambam MedicalCenter conducted an additional comparative study (Fig. 3). This center isqualified to provide this service (ISO 9001: 2000, registration no.IL-30578). Samples were taken from sera of the type 1 diabetes Abs pro-gram, cycle V (January 2009) with RCPA quality assurance.

Determination of anti-chemokines Ab titer

A direct ELISA has been used to determine the chemokine autoantibodytiter in human subjects. Commercially available human purified recombi-nant chemokines (R&D Systems) were coated onto 96-well ELISA plates(Nunc) at concentrations of 20 ng/well. Antisera, in serial dilutions from 2(4) to 2 (29) were added to ELISA plates. Donkey anti-human IgG HRP-conjugated Ab (Jackson Immuno Research Laboratories) was used as de-tection Ab. Abs titer was detected by comparative analysis of sequentialdilutions of sera (cut off Log2Ab titer �9). Results are shown as log2-Abtiter � SE.

Fluid phase radioassay

Serum levels of anti-CCL3 were determined by the method of Vardi et al.(17) as follows: 20 �l of serum was pipet to a tube followed by the 50 �lof assay buffer with or without recombinant human 125I-labeled CCL3(�200,000 cpm/ml; PerkinElmer). The mixture was incubated for 2 h atroom temperature. Then followed the addition of 50 �l of protein and aSepharose incubation for 1 h at room temperature. The precipitate, con-taining bound ligand, was separated by centrifugation at 1500 g for 30 minat 4°C. After aspiration of the supernatant, the precipitate was washed with1.5 ml of ice-cold buffer solution, centrifuged again, and the radioactivitycounted.

Migration assay

THP-1 cells (106) were loaded in the upper chamber of a 6.5-mm diameter,5-�m-pore polycarbonate Transwell culture inserts (Costar). The lowerchamber contained 10 ng/ml CCL3 or CCL2 (R&D Systems), supple-mented with 20 �g IgG purified from a pool of sera from type I diabeticpatients or control subjects (protein G coulomb purification). Cells wereallowed to migrate for 2 h at 37°C in 7.5% CO2 and cells that migratedwere collected and counted using a FACSCalibur (BD Biosciences). Thepercentage of cell migration was calculated as the number of cells thatmigrated to the lower chamber divided by the number of cells originallyplated in the upper chamber.

Determination of autoantibody titer to CIAA, ICA, and GAD

Production of autoantibody titer to CIAA, ICA, and GAD was determinedusing the following commercially available diagnostic kits: Anti-GAD andanti-insulin autoantibodies were tested by radioimmunoassay using a kitpurchased from CIS Bio International.

ICA autoantibodies were determined by a standard immunofluorescencemethod using sections of frozen human group O pancreas (16, 29). End-point dilution titers were examined for the positive samples, and the resultswere expressed in Juvenile Diabetes Foundation units. Titers were con-verted to Juvenile Diabetes Foundation units as recommended by the thirdInternational Workshop of Standardization (16, 29).

Statistical analysis

Statistical analysis was done as according to Ref. 31. Average CCL3 spe-cific autoantibody titter (Log2) was analyzed as mean � SD and signifi-cance was determined by Student’s t test. Receiver operating characteristic(ROC) curve for CCL3 autoantibodies (Figs. 2 and 3) is based on thebinomial distribution to obtain upper and lower bounds for 95% confidenceintervals surrounding estimates of test sensitivity and specificity. Whenstudy results indicated perfect (100%) sensitivity or perfect (100%) spec-ificity in a subgroup of interest, we report the lower 95% confidence bound.Otherwise, we report a symmetrical 95% confidence intervals.

ResultsNewly diagnosed patient with T1DM developed a selectiveautoantibody titer to CCL3 (MIP-1�)

In the first part of our research, we evaluated the presence of aselective autoantibody response to various chemokines that havebeen implicated with inflammatory autoimmunity, includingMIP-1� (CCL3), MCP-1 (CCL2), MIP-1� (CCL4), IL-8

FIGURE 1. T1DM subjects display a selective autoantibody titer toCCL3. A, Sera from 10 subjects diagnosed as new onset T1DM were eval-uated for autoantibody titer to CXCL10, CXCL8, CCL2, CCL3, and CCL4.Results are show as log2Ab titer of individual subjects. B, Pooled sera fromthe above subjects (only IgG) was determines for their ability to inhibitTHP-1 migration induced by CCL3 (b–d) or CCL2 (f–h) as follows: a ande, cells clone alone; b, THP-1 � CCL3; c, THP-1 � CCL3 � IgG fromcontrol (Cont.) subjects; d, THP-1 � CCL3 � IgG from T1DM subjects;f, THP-1 � CCL2; g, THP-1 � CCL2 � IgG from control subjects; h,THP-1 � CCL2 � IgG from T1DM subjects. Results are shown as che-motaxic index � SE of triplicates.

8105The Journal of Immunology

(CXCL8), and IP-10 (CXCL10). Fig. 1A shows that 9 of 10 newlydiagnosed patients with T1DM mount a selective autoantibody ti-ter to CCL3, but not to each of the other chemokines ( p � 0.0001).

It has previously been shown that neutralization of CCL3 effec-tively suppresses spontaneously developed T1DM in NOD mice(25). Our working hypothesis is that selective autoimmunity toCCL3 might participate in the regulation of disease in T1DM pa-tients. In an attempt to further explore this hypothesis we havechecked whether autoantibodies produced in these patients selec-tively neutralizes CCL3. Thus, purified IgG from a pool of serafrom these patients and from the control subjects were analyzed fortheir ability to inhibit CCL3- and CCL2-induced migration of hu-man THP1 cells in a Transwell system. Fig. 1B shows that serafrom T1DM patients, but not from control subjects, could signif-icantly inhibit CCL3-induced migration (Fig. 3, D compared withB and C, p � 0.0001) but not CCL2-induced migration (Fig 3, Hcompared with G and F).

The results of our initial screening (Fig. 1A) motivated us toenlarge our study and determine whether anti-CCL3 Ab produc-tion could be used as a biomarker to distinguish newly diagnosedpatients with T1DM. We therefore compared anti-CCL3 Ab titer insera of 87 newly diagnosed patients with T1DM with 54 age-matched healthy subjects. Fig. 2A shows that mean Log2Ab titer toCCL3 in newly diagnosed patients with T1DM reached the level of12.24 � 0.14, which significantly differed ( p � 0.0001) fromthose recorded in control subjects (7.75 � 0.25). Based on theseobservations, we decided to set up the cut-off titer of Log2Ab titer�9 as positive and analyzed our data accordingly, either with noage limitation (Fig. 2C) or with age limitation of up to18 years ofage, including all the newly diagnosed subjects in the study (Fig.

2D). According to a cut-off �9, ROC curve shows 87.4% sensi-tivity and 94.4% specificity for age-independent analysis (Fig. 2C)and 91.82% sensitivity with 100% specificity when limiting thestudy to patients up to 18 years of age.

An additional set of experiments included analysis of a blindedseries of samples conducted independently by the clinical servicelaboratory of endocrinology at Rambam Medical Center that isqualified to provide this service (ISO 9001: 2000, registration no.IL-30578). Samples were taken from sera of the type 1 diabetesAbs program, cycle V (January 2009) with RCPA quality assur-ance. All analyses were conducted by this service center indepen-dently. In these experiments, sera samples from 30 newly diag-nosed type 1 diabetes patients and age-matched healthy controlswere analyzed for the appearance of anti-CCL3 Abs using threedifferent methods: measuring of Log2Ab titer following serial seradilutions (Fig. 3, A and F), determining the actual OD at threedifferent fixed dilutions (1:500, 1:1000, and 1:2000, Fig. 3, B andG, C and H, and D and I, respectively), and fluid phase radioassay(Fig. 3, E and J). All tests showed a highly significant difference( p � 0.0001) in the sensitivity of anti-CCL3 Ab production be-tween T1DM subjects and healthy controls (Fig. 3).

Autoantibody production to CCL3 in T1DM is disease specific

We have compared the development of autoantibodies to CCL3between 34 subjects suffering from prolonged T1DM (�5 years),87 patients diagnosed as new onset T1DM, 12 subjects sufferingfrom RA and 14 JRA patients, all in comparison with 54 controlsubjects (Fig. 4A). Possible development of autoantibody titerto TNF-1� was also recorded (Fig. 4B). Fig. 4A shows thatanti-CCL3 Ab production is diseases specific, thus while the vast

FIGURE 2. Distribution of auto-antibodies to CCL3 in newly diag-nosed TIDM subjects. A, Meanlog2Ab titer to CCL3 in 87 newly di-agnosed T1DM compared with 54control subjects. Results are shown asmean log2Ab titer � SD (p �0.0001). B, Distribution of autoanti-body titer (Log2) within each group.C and D, ROC curves analysis forsensitivity and specificity of anti-CCL3 autoantibodies T1DM subjectsin 87 newly diagnosed T1DM sub-jects and 54 controls. C, Analysis ofall samples. D, Age limitation to�18. AUC, area under the curve.

8106 SELECTIVE AUTOANTIBODY PRODUCTION AGAINST CCL3

majority of newly onset T1DM patients and subjects sufferingfrom prolonged T1DM (87.4% and 70.6%, respectively) displayeda �log29 titer to CCL3, only 14.3% and 16.7% of JRA and RApatients, respectively, were anti-CCL3 positive ( p � 0.01). Inter-estingly, T1DM patients did not display any notable Ab titer to

TNF-�, which was a hallmark for the diagnosis of RA (1), as wellas JRA (Fig. 4B), implicating, once again, the selectivity in thebreakdown of tolerance during autoimmunity.

As a further control, we looked for a disease in which pancreatic�-cell function is also altered at an early age. Thus, CF with pan-creatic insufficiency (32), in which pancreatic �-cell function isaltered, leading to glucose intolerance and diabetes (33), was se-lected as a comparative disease for T1DM. We have comparedautoantibody production to CCL3 in sera of 10 CF patients withpancreatic insufficiency and impaired glucose tolerance, whichwere all negative for autoantibody production to either ICA orGAD (Fig. 4C). Nine of them displayed low autoantibody produc-tion to CCL3, which was comparable to control age-matchedhealthy subjects, and only one patient displayed a significant titerto CCL3 (log211). Statistical analysis of both groups showed nosignificant differences (Log2 � SE of 8.1 � 0.43 compared with7.6 � 0.3). It should be noticed that two other patients with CF andpancreatic insufficiency were excluded from the study becausethey were found to be positive to ICA (one also to GAD). Bothdisplayed a significant titer to CCL3 (log211). Of 10 patients thatwere identified as CF without pancreatic insufficiency and werenegative to ICA or GAD, one patient displayed a significant titerto CCL3 (log210) (not shown). Thus, alteration of the �-cell func-tion resulting in glucose intolerance is not sufficient for the gen-eration of anti-CCL3 autoantibody production.

Frequency of anti-CCL3 Ab production in T1DM subjects ishigher than in other known targets currently serving asbiomarkers

The current diagnosis of T1DM is based on measuring autoanti-bodies to CIAA, ICA, and GAD (15–17). We have compared au-toantibody production to CIAA, ICA, GAD, and CCL3 in 87newly diagnosed T1DM (Fig. 5A). Although 87.4% of these sub-jects were positive (cut-off �9) for anti-CCL3, only 63% (54/86),

FIGURE 3. Comparative analysis of autoantibody production to CCL3 as determined by different techniques. Comparative analysis of autoantibodyproduction to CCL3 in sera of T1DM patients and control subjects as determined by measuring Log2Ab titer following serial sera dilutions (A and F),determining the actual OD at three different fixed dilutions (B and G, 1/500; C and H, 1/1000; and D and I, 1/2000), and fluid phase radioassay, determinedin cpm (E and J). Panels A–E show individual levels, whereas panels F–J display ROC curves analysis for sensitivity and specificity of anti-CCL3autoantibodies. AUC, area under the curve.

FIGURE 4. Autoantibodies production to CCL3 is disease selective. A,Comparative analysis of autoantibody production to CCL3 (cut off Log2Abtiter � 9) of subjects with prolonged T1DM (70.6%), new onset T1DM(87.4%), RA (16.7%), and JRA (14.3%), compared with control subjects(5.6%). B, Mean log2Ab titer to TNF-� � SE as determined in sera sam-ples specified in A. C, Distribution of autoantibody titer (Log2) of anti-CCL3 Ab production in sera of CF patients with glucose intolerance andcontrol subjects. AUC, area under the curve.

8107The Journal of Immunology

60% (50/83), and 48% (39/82) of them were positive for GAD,ICA, and CIAA autoantibody production ( p � 0.01 for the com-parison of anti-CCL3 to each of the others), respectively. Our dataanalysis shows that a combined test of all three biomarkers (GAD,ICA, and CIAA) would cover �85% of patients.

Anti-CCL3 Abs as a biomarker for diabetes in prediabeticsubjects

We have examined whether anti-CCL3 also appears in prediabeticsubjects (first-degree relatives of patients with T1DM with positiveautoantibodies of at least one of the following: anti-Gad (85%),ICA (70%), and anti-insulin Abs (90%). At the prediabetic stage,19 of these 20 subjects (95%) were anti-CCL3 positive.

Finally, 18 first-degree relatives of patients with T1DM, whowere autoantibody negative to CIAA, ICA, or GAD, were exam-ined for autoantibody production to CCL3 (measured by OD at adilution of 1:500). Of these subjects, only two (11%) were anti-CCL3 positive. We are monitoring these children for possible fu-ture development of autoantibody response to CIAA, ICA, orGAD, as well as for clinical signs of T1DM.

DiscussionWe have previously shown that targeted DNA vaccines encodinginflammatory cytokines and chemokines could be used to rapidlysuppress ongoing inflammatory autoimmunity, and that diseasesuppression is due to an accelerated autoantibody production to thegene products of each vaccine (3, 34–37). We have then showedthat the rapid effect of this way of therapy is due to amplificationof an ongoing beneficial response that participates in the naturalregulation of disease. It is possible, though hard to be proven, thatthis response is also helpful for human subjects suffering from RA,which makes it a possible target for beneficial amplification (1).Based on this study, we have established a target discovery plat-form aimed at identifying key mediators of different inflammatoryautoimmune disease in humans.

Of the 50 known chemokines, the CC chemokines, mostlyMIP-1� (CCL3), MCP-1 (CCL2), and MIP-1� (CCL4), as well asthe CXC chemokines IL-8 (CXCL8) and IP-10 (CXCL10), weremostly defined as inducers of the inflammatory process in variousinflammatory autoimmune diseases (38–43). The CC chemokinesRANTES (CCL5) was also associated with RA (44–46). Of thesechemokines, the vast majority of T1DM subjects produce neutral-izing Abs exclusively to CCL3 (Fig. 1) but not others, particularlyCCL2, which has been well implicated with several autoimmunediseases like multiple sclerosis, RA, myocarditis, and their exper-imental models (47–49). One possibility is that the inflammatory

process in T1DM is regulated differently than other T cell-medi-ated autoimmune diseases, like RA, multiple sclerosis, and myo-carditis. Indeed, Cameron et al. have shown that from the CCchemokines selective neutralization of CCL3 suppresses type 1diabetes in NOD mice (25). Subsequently, it has been shown thatthe relatives at risk of developing T1DM are associated with up-regulation of CCL3 (MIP 1�) and CCL4 (MIP1�), accompaniedby down-regulation of CCL2 (MCP1). These findings suggest op-posed functions of these chemokines in the disease process (50). Itshould be noted that vast majority of our RA and JRA patients aresubjected to a low level of steroid therapy, which does not termi-nate their ability to spontaneously generate anti-TNF-� Ab titer(Fig. 4B). Interestingly, T1DM subjects do not mount any autoan-tibody titer to this inflammatory cytokine (Fig. 4B).

It has previously been shown that neutralization of CCL3 effec-tively suppresses spontaneously developed T1DM in NOD mice(25). In this paper, we show that autoantibodies produced inT1DM subjects are neutralizing Abs, and speculate that they mightparticipate in the natural regulation of disease. It is tempting tohypothesize that in an attempt to restrain the destructive immuneprocess, the immune system would produce an autoantibodyagainst one of the key mediators involved in the disease processand that CCL3 would be one of the strongest candidates, thoughthe exact mechanism by which the immune system does so is stillelusive. From the therapeutic perspective, this may suggest CCL3as a favorable target for therapy. From the diagnostically orientedperspective, according to our data, anti-CCL3 Abs were positive in�87% of patients, whereas only 63% (54/86), 60% (50/83), and48% (39/82) were positive for anti-GAD, ICA and CIAA, respec-tively. Our data analysis shows that a combined test of all threebiomarkers (GAD, ICA, and CIAA) would cover �85% of pa-tients. Therefore an additional anti-CCL3 test will increase theaccuracy and reliability of T1DM diagnosis. Moreover, anti-CCL3is positive in prediabetic patients and continues to be positive sev-eral years after the diagnosis of diabetes.

Finally, as emerging therapies for T1DM are coming, an earlydiagnostic of disease, even during its preclinical stage, would becritical for successful therapy. We show that 19 of 20 subjects(95%) who were at prediabetic stage displayed an autoantibodyresponse to CCL3. We therefore believe that our study is mostlyimportant for an early diagnosis of disease and its therapeuticimplications.

DisclosuresN.K., N.S., and G.W. hold a patent on detection of T1DM by anti-CCL3Abs that has been licensed out to Micro Medic (Israel).

References1. Wildbaum, G., M. Nahir, and N. Karin. 2003. Beneficial autoimmunity to proin-

flammatory mediators restrains the consequences of self-destructive immunity.Immunity 19: 679–688.

2. Feldmann, M., F. M. Brennan, and R. N. Maini. 1996. Role of cytokines inrheumatoid arthritis. Annu. Rev. Immunol. 14: 397–440.

3. Wildbaum, G., S. Youssef, and N. Karin. 2000. A targeted DNA vaccine aug-ments the natural immune response to self TNF-� and suppresses ongoing ad-juvant arthritis. J. Immunol. 165: 5860–5866.

4. Castano, L., and G. S. Eisenbarth. 1990. Type-I diabetes: a chronic autoimmunedisease of human, mouse, and rat. Annu. Rev. Immunol. 8: 647–679.

5. Hutchings, P., H. Rosen, L. O’Reilly, E. Simpson, S. Gordon, and A. Cooke.1990. Transfer of diabetes in mice prevented by blockade of adhesion-promotingreceptor on macrophages. Nature 348: 639–642.

6. Kawasaki, E., H. Takino, M. Yano, S. Uotani, K. Matsumoto, Y. Takao,Y. Yamaguchi, S. Akazawa, and S. Nagataki. 1994. Autoantibodies to glutamicacid decarboxylase in patients with IDDM and autoimmune thyroid disease. Di-abetes 43: 80–86.

7. Pleau, J. M., F. Fernandez-Saravia, A. Esling, F. Homo-Delarche, andM. Dardenne. 1995. Prevention of autoimmune diabetes in nonobese diabeticfemale mice by treatment with recombinant glutamic acid decarboxylase (GAD65). Clin. Immunol. Immunopathol. 76: 90–95.

FIGURE 5. Comparative analysis of anti-CCL3 autoantibodies to otherbiomarkers for T1DM diagnosis. A, Comparative analysis of anti-CCL3autoantibodies (cut off Log2Ab titer � 9) to other biomarkers for T1DMdiagnosis including CIAA (48%), ICA (60%), and GAD (63%). B, Com-parative analysis of anti-CCL3 autoantibodies (cut off Log2Ab titer � 9)between prediabetic subjects and new onset T1DM (87.4%).

8108 SELECTIVE AUTOANTIBODY PRODUCTION AGAINST CCL3

8. Rudy, G., N. Stone, L. C. Harrison, P. G. Colman, P. McNair, V. Brusic,M. B. French, M. C. Honeyman, B. Tait, and A. M. Lew. 1995. Similar peptidesfrom two � cell autoantigens, proinsulin and glutamic acid decarboxylase, stim-ulate T cells of individuals at risk for insulin-dependent diabetes. Mol. Med. 1:625–633.

9. Atkinson, M. A. 2000. The $64000 question in diabetes continues. Lancet356: 4–6.

10. Lohmann, T., M. Hawa, R. D. Leslie, R. Lane, J. Picard, and M. Londei. 2000.Immune reactivity to glutamic acid decarboxylase 65 in stiffman syndrome andtype 1 diabetes mellitus. Lancet 356: 31–35.

11. Scofield, R. H. 2004. Autoantibodies as predictors of disease. Lancet 363:1544–1546.

12. Sellers, E., G. Eisenbarth, T. K. Young, and H. J. Dean. 2000. Diabetes-associ-ated autoantibodies in aboriginal children. Lancet 355: 1156.

13. Kaufman, D. L., M. G. Erlander, M. Claire-Salzler, M. A. Atkinson,N. K. Maclaren, and A. J. Tobin. 1992. Autoimmunity to two forms of glutaminedecarbosylase in insulin-dependent diabetes mellitus. J. Clin. Invest. 89: 283–292.

14. Armstrong, N. W., and D. B. Jones. 1994. Epitopes of GAD 65 in insulin-de-pendent diabetes mellitus. Lancet 344: 406–407.

15. Atkinson, M. A., M. A. Bowman, L. Campbell, B. L. Darrow, D. L. Kaufman,and N. K. Maclaren. 1994. Cellular immunity to a determinant common to glu-tamate decarboxylase and coxsackie virus in insulin-dependent diabetes. J. Clin.Invest. 94: 2125–2129.

16. Schatz, D., J. Krischer, G. Horne, W. Riley, R. Spillar, J. Silverstein, W. Winter,A. Muir, D. Derovanesian, S. Shah, et al. 1994. Islet cell antibodies predictinsulin-dependent diabetes in United States school age children as powerfully asin unaffected relatives. J. Clin. Invest. 93: 2403–2407.

17. Vardi, P., S. A. Dib, M. Tuttleman, J. E. Connelly, M. Grinbergs, A. Radizabeh,W. J. Riley, N. K. Maclaren, G. S. Eisenbarth, and J. S. Soeldner. 1987. Com-petitive insulin autoantibody assay: prospective evaluation of subjects at high riskfor development of type I diabetes mellitus. Diabetes 36: 1286–1291.

18. Zlotnic, A., and O. Yoshei. 2000. Chemokines: a new classification system andtheir role in immunity. Immunity 12: 121–127.

19. Mackay, C. R. 2001. Chemokines: immunology’s high impact factors. Nat. Im-munol. 2: 95–101.

20. Rollins, B. J. 1997. Chemokines. Blood 90: 909–928.21. Proudfoot, A. E. 2002. Chemokine receptors: a multifaceted therapeutic targets.

Nat. Rev. Immunol. 2: 106–115.22. Christen, U. 2007. Chemokines as drug targets in type 1 diabetes. Endocr. Metab.

Immune Disord. Drug Targets 7: 7–12.23. Christen, U., D. B. McGavern, A. D. Luster, M. G. von Herrath, and

M. B. Oldstone. 2003. Among CXCR3 chemokines, IFN-�-inducible protein of10 kDa (CXC chemokine ligand (CXCL) 10) but not monokine induced by IFN-�(CXCL9) imprints a pattern for the subsequent development of autoimmune dis-ease. J. Immunol. 171: 6838–6845.

24. Frigerio, S., T. Junt, B. Lu, C. Gerard, U. Zumsteg, G. A. Hollander, and L. Piali.2002. � Cells are responsible for CXCR3-mediated T-cell infiltration in insulitis.Nat. Med. 8: 1414–1420.

25. Cameron, M. J., G. A. Arreaza, M. Grattan, C. Meagher, S. Sharif,M. D. Burdick, R. M. Strieter, D. N. Cook, and T. L. Delovitch. 2000. Differentialexpression of CC chemokines and the CCR5 receptor in the pancreas is associ-ated with progression to type I diabetes. J. Immunol. 165: 1102–1110.

26. Palacios, I., M. J. Lopez-Armada, P. Hernandez, O. Sanchez-Pernaute,S. Gutierrez, R. Miguelez, J. Martinez, J. Egido, and G. Herrero-Beaumont. 1998.Tenidap decreases IL-8 and monocyte chemotactic peptide-1 (MCP-1) mRNAexpression in the synovial tissue of rabbits with antigen arthritis and in culturedsynovial cells. Clin. Exp. Immunol. 111: 588–596.

27. Bradley, L. M., V. C. Asensio, L. K. Schioetz, J. Harbertson, T. Krahl,G. Patstone, N. Woolf, I. L. Campbell, and N. Sarvetnick. 1999. Islet-specificTh1, but not Th2, cells secrete multiple chemokines and promote rapid inductionof autoimmune diabetes. J. Immunol. 162: 2511–2520.

28. Szalai, C., A. Csaszar, A. Czinner, T. Szabo, P. Panczel, L. Madacsy, andA. Falus. 1999. Chemokine receptor CCR2 and CCR5 polymorphisms in childrenwith insulin-dependent diabetes mellitus. Pediatr. Res. 46: 82–84.

29. Boitard, C., E. Bonifacio, G. F. Bottazzo, H. Gleichmann, and J. Molenaar. 1988.Immunology and Diabetes Workshop: report on the Third International (Stage 3)Workshop on the Standardisation of Cytoplasmic Islet Cell Antibodies: Held inNew York, New York, October 1987. Diabetologia 31: 451–452.

30. Karjalainen, J., P. Salmela, J. Ilonen, H. M. Surcel, and M. Knip. 1989. A com-parison of childhood and adult type I diabetes mellitus. N. Engl. J. Med. 320:881–886.

31. Leman, E. S., R. E. Schoen, J. L. Weissfeld, G. W. Cannon, L. J. Sokoll,D. W. Chan, and R. H. Getzenberg. 2007. Initial analyses of colon cancer-specificantigen (CCSA)-3 and CCSA-4 as colorectal cancer-associated serum markers.Cancer Res. 67: 5600–5605.

32. Brennan, A. L., D. M. Geddes, K. M. Gyi, and E. H. Baker. 2004. Clinicalimportance of cystic fibrosis-related diabetes. J. Cyst. Fibros. 3: 209–222.

33. Bismuth, E., K. Laborde, P. Taupin, G. Velho, V. Ribault, F. Jennane, E. Grasset,I. Sermet, J. de Blic, G. Lenoir, and J. J. Robert. 2008. Glucose tolerance andinsulin secretion, morbidity, and death in patients with cystic fibrosis. J. Pediatr.152: 540–545.

34. Youssef, S., G. Wildbaum, G. Maor, N. Lanir, A. Gour-Lavie, N. Grabie, andN. Karin. 1998. Long-lasting protective immunity to experimental autoimmuneencephalomyelitis following vaccination with naked DNA encoding C-C chemo-kines. J. Immunol. 161: 3870–3879.

35. Salomon, I., N. Netzer, G. Wildbaum, S. Schif-Zuck, G. Maor, and N. Karin.2002. Targeting the function of IFN-�-inducible protein 10 suppresses ongoingadjuvant arthritis. J. Immunol. 169: 2685–2693.

36. Wildbaum, G., N. Netzer, and N. Karin. 2002. Plasmid DNA encoding IFN-�-inducible protein 10 redirects antigen-specific T cell polarization and suppressesexperimental autoimmune encephalomyelitis. J. Immunol. 168: 5885–5892.

37. Goldberg, R., G. Wildbaum, Y. Zohar, G. Maor, and N. Karin. 2004. Suppressionof ongoing adjuvant-induced arthritis by neutralizing the function of the p28subunit of IL-27. J. Immunol. 173: 1171–1178.

38. Manczak, M., S. Jiang, B. Orzechowska, and G. Adamus. 2002. Crucial role ofCCL3/MIP-1� in the recurrence of autoimmune anterior uveitis induced withmyelin basic protein in Lewis rats. J. Autoimmun. 18: 259–270.

39. dos Santos, A. C., M. M. Barsante, R. M. Arantes, C. C. Bernard, M. M. Teixeira,and J. Carvalho-Tavares. 2005. CCL2 and CCL5 mediate leukocyte adhesion inexperimental autoimmune encephalomyelitis: an intravital microscopy study.J. Neuroimmunol. 162: 122–129.

40. Badolato, R., A. N. Ponzi, M. Millesimo, L. D. Notarangelo, and T. Musso. 1997.Interleukin-15 (IL-15) induces IL-8 and monocyte chemotactic protein 1 produc-tion in human monocytes. Blood 90: 2804–2809.

41. Karpus, W. J., and K. J. Kennedy. 1997. MIP-1� and MCP-1 differentially reg-ulate acute and relapsing autoimmune encephalomyelitis as well as Th1/Th2 lym-phocyte differentiation. J. Leukocyte Biol. 62: 681–687.

42. Plater-Zyberk, C., A. J. Hoogewerf, A. E. Proudfoot, C. A. Power, andT. N. Wells. 1997. Effect of a CC chemokine receptor antagonist on collageninduced arthritis in DBA/1 mice. Immunol. Lett. 57: 117–120.

43. Qin, S., J. B. Rottman, P. Myers, N. Kassam, M. Weinblatt, M. Loetscher,A. E. Koch, B. Moser, and C. R. Mackay. 1998. The chemokine receptorsCXCR3 and CCR5 mark subsets of T cells associated with certain inflammatoryreactions. J. Clin. Invest. 101: 746–754.

44. Barnes, D. A., J. Tse, M. Kaufhold, M. Owen, J. Hesselgesser, R. Strieter,R. Horuk, and H. D. Perez. 1998. Polyclonal antibody directed against humanRANTES ameliorates disease in the Lewis rat adjuvant-induced arthritis model.J. Clin. Invest. 101: 2910–2919.

45. Youssef, S., G. Maor, G. Wildbaum, N. Grabie, A. Gour-Lavie, and N. Karin.2000. C-C chemokine-encoding DNA vaccines enhance breakdown of toleranceto their gene products and treat ongoing adjuvant arthritis. J. Clin. Invest. 106:361–371.

46. Pierer, M., J. Rethage, R. Seibl, R. Lauener, F. Brentano, U. Wagner,H. Hantzschel, B. A. Michel, R. E. Gay, S. Gay, and D. Kyburz. 2004. Chemo-kine secretion of rheumatoid arthritis synovial fibroblasts stimulated by Toll-likereceptor 2 ligands. J. Immunol. 172: 1256–1265.

47. Goser, S., R. Ottl, A. Brodner, T. J. Dengler, J. Torzewski, K. Egashira,N. R. Rose, H. A. Katus, and Z. Kaya. 2005. Critical role for monocyte che-moattractant protein-1 and macrophage inflammatory protein-1� in induction ofexperimental autoimmune myocarditis and effective anti-monocyte chemoattrac-tant protein-1 gene therapy. Circulation 112: 3400–3407.

48. Ohta, M., Y. Kitadai, S. Tanaka, M. Yoshihara, W. Yasui, N. Mukaida,K. Haruma, and K. Chayama. 2002. Monocyte chemoattractant protein-1 expres-sion correlates with macrophage infiltration and tumor vascularity in humanesophageal squamous cell carcinomas. Int. J. Cancer 102: 220–224.

49. Ogata, H., M. Takeya, T. Yoshimura, K. Takagi, and K. Takahashi. 1997. Therole of monocyte chemoattractant protein-1 (MCP-1) in the pathogenesis of col-lagen-induced arthritis in rats. J. Pathol. 182: 106–114.

50. Hanifi-Moghaddam, P., S. Kappler, J. Seissler, S. Muller-Scholze, S. Martin,B. O. Roep, K. Strassburger, H. Kolb, and N. C. Schloot. 2006. Altered chemo-kine levels in individuals at risk of Type 1 diabetes mellitus. Diabet. Med. 23:156–163.

8109The Journal of Immunology