Sle Review

download Sle Review

of 13

Transcript of Sle Review

  • 7/30/2019 Sle Review

    1/13

  • 7/30/2019 Sle Review

    2/13

    Aberrant Phenotype and Function of Myeloid Dendritic Cells

    in Systemic Lupus Erythematosus1

    Dacheng Ding, Hemal Mehta, W. Joseph McCune, and Mariana J. Kaplan2

    Systemic lupus erythematosus (SLE) is characterized by a systemic autoimmune response with profound and diverse T cell

    changes. Dendritic cells (DCs) are important orchestrators of immune responses and have an important role in the regulation of

    T cell function. The objective of this study was to determine whether myeloid DCs from individuals with SLE display abnormal-

    ities in phenotype and promote abnormal T cell function. Monocyte-derived DCs and freshly isolated peripheral blood myeloid

    DCs from lupus patients displayed an abnormal phenotype characterized by accelerated differentiation, maturation, and secretion

    of proinflammatory cytokines. These abnormalities were characterized by higher expression of the DC differentiation marker

    CD1a, the maturation markers CD86, CD80, and HLA-DR, and the proinflammatory cytokine IL-8. In addition, SLE patients

    displayed selective down-regulation of the maturation marker CD83 and had abnormal responses to maturation stimuli. These

    abnormalities have functional relevance, as SLE DCs were able to significantly increase proliferation and activation of allogeneic

    T cells when compared with control DCs. We conclude that myeloid DCs from SLE patients display significant changes in

    phenotype which promote aberrant T cell function and could contribute to the pathogenesis of SLE and organ damage. The

    Journal of Immunology, 2006, 177: 58785889.

    Systemic lupus erythematosus is an autoimmune disease of

    unclear etiology that affects multiple organs. Individuals

    with systemic lupus erythematosus (SLE)3 present with

    numerous and diverse immune system abnormalities and display a

    prominent imbalance in different T cell subset functions (13).

    Indeed, individuals with SLE have autoreactive T cells that are

    specific for ubiquitous self-peptides and provide pathogenic B cell

    help (4, 5). A pan T cell dysfunction in SLE is characterized by

    exaggerated CD4 T cell responses (6, 7) and diminished CD8 T

    cell activities (8). Other alterations include changes in proliferative

    T cell responses as well as increased expression of early and late

    T cell activation markers (3, 7, 911). In addition, alterations inTh1 and/or Th2 lymphocyte function have been described (1216),

    resulting in production of cytokines that up-regulate autoantibody

    production by B cells, promote immune complex formation, and

    increase the apoptotic load (17, 18). The exact role that accessory

    cells might have in inducing abnormal T cell responses in SLE is

    unclear; however, different groups have proposed that T cell dis-

    turbances in SLE could be induced or promoted, at least in part, by

    alterations in dendritic cell (DC) phenotype and function, because

    these are key regulators of the immune system (1922). DCs com-

    prise several subsets with different stages of maturation (20). Im-

    mature, Ag-capturing DCs sense diverse signals that induce them

    to undergo a maturation process, whose hallmarks are the up-reg-

    ulation of the molecule CD83, as well as of a number of costimu-

    latory and adhesion molecules, migration into lymphoid organs

    (23), and acquisition of the capacity to activate quiescent, naive,

    and memory lymphocytes (20, 2430). DCs can induce either T

    cell tolerance or strong innate and adaptive immunity to specific

    Ag. In general, tolerance is initiated when DCs are immature,

    whereas the initiation of immunity requires an effective DC mat-

    uration signal. After reaching the lymph nodes, DCs present pro-

    cessed Ag to T cells via both classical (class I and II MHC) andnonclassical (CD1 family) pathways (24, 31, 32). In addition to

    their maturation status, DCs can be subdivided into a CD11c

    (plasmacytoid DC) and a CD11c (myeloid DC) subset (20, 33).

    The CD11c subset follows a myeloid differentiation pathway in

    which monocytes serve as precursors. These DC subsets have dif-

    ferent migration programs and very likely have divergent roles in

    the induction and regulation of the immune response (33, 34)

    In the past few years, significant progress has been made in

    elucidating the important role that IFN--producing plasmacytoid

    DCs have in promoting autoimmune responses in SLE (22, 35, 36).

    Much less is known about the role that myeloid DCs play in con-

    tributing to the pathogenesis of SLE. Indeed, it is unclear whether

    myeloid DCs present phenotypic and functional changes that may

    promote the autoimmune features of lupus and whether such dif-

    ferences might potentiate aberrant cognate interactions with T

    cells. We now report that myeloid DCs of individuals with SLE

    display evidence of aberrant phenotype and function which may

    contribute to the T cell abnormalities described in this disease.

    Materials and MethodsPatient selection

    Patients with SLE and rheumatoid arthritis (RA) fulfilled the AmericanCollege of Rheumatology criteria for these diseases (3739) and were re-cruited from the outpatient rheumatology clinic and inpatient services atthe University of Michigan, and from the Michigan Lupus Cohort (AnnArbor, MI). Healthy controls were obtained by advertisement. SLE activity

    Department of Internal Medicine, Division of Rheumatology, University of Michigan,Ann Arbor, MI 48109

    Received for publication June 15, 2005. Accepted for publication August 12, 2006.

    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 This work was supported by Public Health Service Grants AR050554 andAR048235 and by the Anthony S. Gramer Fund in Inflammation Research (all toM.J.K.). D.D. was supported by Training Grant T32 AR 07080. W.J.M. was sup-ported by the Herb and Carol Amster Lupus Research Fund and the Klein LupusResearch Fund. This research was also supported (in part) by the National Institutesof Health through University of Michigans Cancer Center Support Grant (P30CA46592) and the Rheumatic Diseases Core Center Grant (P30 AR48310).

    2 Address correspondence and reprint requests to Dr. Mariana J. Kaplan, Division ofRheumatology, University of Michigan, 5520 MSRBI, 1150 West Medical CenterDrive, Ann Arbor, MI 48109-0680. E-mail address: [email protected]

    3 Abbreviations used in this paper: SLE, systemic lupus erythematosus; DC, dendriticcell; RA, rheumatoid arthritis; SLEDAI, SLE disease activity index; 6-MP, 6-mer-captopurine; MMF, mycophenolate-mofetil.

    The Journal of Immunology

    Copyright 2006 by The American Association of Immunologists, Inc. 0022-1767/06/$02.00

    p

    ,

    j

    g

    http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/
  • 7/30/2019 Sle Review

    3/13

    was assessed by the SLE disease activity index (SLEDAI) (40). Patient andcontrol cells were paired and studied in parallel. Overall, 146 SLE patients, 76healthy controls and 35 RA patients were studied. Information regarding thedemographics, disease activity, and use of medications is provided in Table I.

    Reagents

    Human rIL-4, TNF-, and IL-2 were purchased from PeproTech. HumanGM-CSF was a gift from Berlex. rIFN-2b was obtained from Schering.X-vivo 10 serum-free medium was from BioWhittaker. LPS and PHA werepurchased from Sigma-Aldrich. The following anti-human mAbs conju-

    gated to FITC, PE, CyChrome, and allophycocyanin were used: anti-CD1a,CD3, CD4, CD8, CD11c, CD14, CD25, CD40L, CD69, CD80, CD83,CD86, HLA-DR, and isotype controls (all obtained from BD Biosciences).The pan T cell isolation kit was obtained from Miltenyi Biotec. CFSE wasobtained from Molecular Probes.

    Generation of monocyte-derived DCs

    Myeloid DCs were generated from human peripheral monocytes as previ-ously described (41). In brief, human PBMC were separated by standarddensity gradient centrifugation on Ficoll-Hypaque Plus (Amersham Bio-sciences) and resuspended at 6 106 cells/ml in RPMI 1640 with L-glutamine and 10% FBS. The cells were transferred to tissue culture platesand allowed to adhere to the plastic surface for 1 h at 37C. Nonadherentcells were then removed by washing with PBS. Monocyte recovery rateafter adherence was 89% in both healthy controls and SLE patients. The

    adhered monocytes were further cultured for 57 days in DC inductionmedium (serum-free X-vivo-10 containing 20 ng/ml GM-CSF and 510ng/ml IL-4). In some experiments, cells were further purified using metri-zamide gradient (Sigma-Aldrich). The purity of myeloid DCs obtainedunder our experimental conditions was 90%, as confirmed by flow cy-tometric analysis (data not shown). At days 57, cells were harvested foranalysis or stimulated with DC maturation stimuli (41) (0.52 g/ml LPSand/or 10100 ng/ml TNF-) for 48 h. In additional experiments, freshlyisolated monocytes were cultured for 7 days with GM-CSF, with or withoutIL-4, in the presence or absence of 100 or 1000 U of rIFN-.

    Immunofluorescence staining, FACS, and fluorescent microscopy

    analysis

    DCs were washed with PBS/0.2% BSA and FcRs were blocked by incu-bating cells for 20 min in PBS with 40% control human sera or with anti-FcAb (Miltenyi Biotec). Cells were then incubated for 30 min at 4C with

    0.060.15 g/ml fluorochrome-conjugated mAb following the manufac-turers directions. Cells were washed three times with PBS/0.2% BSA,fixed in 1% paraformaldehyde and analyzed in a FACSCalibur flow cy-tometer (BD Biosciences), using previously described protocols. Data anal-ysis was performed using Lysys II software (BD Biosciences) and Win-MDI 2.8 software ( http://facs.scripps.edu ). Stained cells were gated byside scatter and forward scatter characteristics and further identified bysurface markers. The results were expressed as the percentage of cellsstaining positive for different markers as well as by mean channel fluores-cence. The cutoff point for positive staining was above the level of thecontrol isotype Abs.

    For fluorescent microscopy, DCs were derived from monocytes as statedabove. At day 7, cells were harvested, washed, and stained with the specificmAbs mentioned above to characterize DC phenotype and morphology.Cells were then fixed in 1% formalin in PBS for 30 min, and transferred toa microscope slide using ProLong Antifade kit (Molecular Probes). Slideswere analyzed using a Zeiss LSM 510 META Laser Scanning Microscope(Carl Zeiss Advanced Imaging Microscopy).

    Isolation and characterization of myeloid DCs from peripheral

    blood

    PBMCs were isolated from peripheral blood of SLE patients and controlsby Ficoll gradient as described above. PBMCs were incubated with twodifferent mixtures of Abs: 1) CD11c-FITC, CD1a-allophycocyanin, CD83-PE, and CD86-PE/Cy5, and 2) CD11c-FITC, CD14-allophycocyanin/Cy5,CD86-PE/Cy5 and HLA-DR-allophycocyanin. After incubation for 30 minwith these Abs at 4C, cells were washed, fixed in PBS/1% paraformal-dehyde, and analyzed by FACS by gating the CD11c population andexcluding CD14 cells.

    Drug treatment

    Monocytes were cultured as stated above to induce DC differentiation, inthe presence or absence of graded concentrations of indomethacin (0.011g/ml), hydroxychloroquine (0.022 g/ml), hydrocortisone (0.011 M),6-mercaptopurine (6-MP) (0.011 M) and mycophenolate-mofetil(MMF) (0.044 g/ml; all obtained from Sigma-Aldrich) or vehicle (42

    46). A stock solution of 6-MP was prepared in dimethylformamide at aconcentration of 10 mg/ml. The stock solution was diluted in assay diluent(80% culture medium/20% ethanol) to yield a 6-MP working solution of 80g/ml or less as indicated. The working solution of 6-MP as well as theother materials prepared in assay diluent were then further diluted 1/40 intothe cell cultures for the tests. The final concentrations of ethanol (0.1%) ordimethylformamide (0.02%) do not yield significant effects on the cellcultures (11, 47). Indomethacin was prepared in a concentration of 500 mMin absolute ethanol, then diluted to final concentrations in the cell culturemedium (11). Control cells were treated with an equal volume of the sol-vent. MMF was prepared as previously described (43). At day 7, DCs werewashed and analyzed by flow cytometry for expression of differentiation(CD1a) and maturation (CD83, CD86, HLA-DR) markers.

    RNA extraction and quantitative real-time RT-PCR

    Total RNA was isolated from myeloid DCs using the RNeasy kit with

    DNase I digestion (Qiagen) to remove possible genomic DNA contamina-tion, and reverse transcribed to cDNA using the SuperScript III first-strandsynthesis system (Invitrogen Life Technologies) with Oligo(dT)30 primer.For real-time detection of target and reference gene expression, eight pairprimers and probes were designed as follows: 83 forward (F): 5-CTGCTCCTGAGCTGCGCCTACA; CD83 reverse (R): 5-CACCACCCTCCAATAACTTGAC; CD83 probe: 5-ATCCGCAGGTTCCCTACACGGTCTCC; CD80 F: 5-CTTCAACTGGAATACAACCAAGCA; CD80 R: 5-TGCATCTTGGGGCAAAGCAGTA; CD80 probe: 5-CTCCCATCCTGGGCCATTACCTTAATC; CD1a F: 5-GTCCTCTACTGGGAGCATCACA; CD1a R: 5-GTCTTAACAGAAACAGCGTTTCC; CD1a probe:

    Table I. Demographic and clinical characteristics of human subjects studied

    Variable Lupus Control RA

    Number studied 146 76 35Age(mean range) 44.5 11.5 37 13.5 51 14.4Females n (%) 130 (89) 55 (72.3) 18 (51.5)Males n (%) 16 (11) 21 (27.6) 17 (48.5)

    Lupus disease activity (mean SEM) 4.5 3SLEDAI 2 (%) 77SLEDAI 2 (%) 69

    MedicationsAntimalarials (%) 68.5 40.5Azathioprine (%) 5.47 0Cyclophosphamide (%) 1.36 0Methotrexate (%) 1.36 51.4Mycophenolate mofetil (%) 24.65 0Prednisone (0.5 mg/kg/day) (%) 34.2 51.4Prednisone (0.51 mg/kg/day) (%) 9.5 5.7Prednisone (1 mg/kg/day) (%) 4.1 8.5

    5879The Journal of Immunology

    p

    ,

    j

    g

    http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/
  • 7/30/2019 Sle Review

    4/13

    5-CTTGGCGGTGATAGTGCCTTTACTTCTT; CD86 F: 5-GACAGGCATTTGTGACAGCACTA; CD86 R: 5-TCT GCA GTC TCA TTGAAA TAA GC; CD86 probe: 5-TTCCTGCTCTCTGGTGCTGCTCCTCT;CD14 F: 5-GGTGCCGCTGTGTAGAAAGAAGC; CD14 R: 5-GGTTCTGGCGTGGTCGCAGAGAC; CD14 probe: 5-TTATCGACCATGGAGCGCGCGT; DRa F: 5-TCAAGGTGCATTGGCCAACATAG; DRaR: 5-CTCTCAGTTCCACAGGGCTGTTC; DRa probe: 5-CGATCACCAATGTACCTCCAGAG; PBGD-F 5-GGCAATGCGGCTGCAA-3 ;PBGD-R 5-GGGTACCCACGCGAATCAC-3 ; PBGD probe 5-CTCATCTTTGGGCTGTTTTCTTCCGCC; -actin F: 5-AGCCTTCCTTC

    CTGGGCATGGA; -actin R: 5-CTCAGGAGGAGCAATGATCTTGA;-actin probe: 5-ACATCCGCAAAGACCTGTACGCCAACA.Probes were labeled with 5-6-FAM/3-TAMRA (Integrated DNA

    Technologies). HotStar Taq polymerase (Qiagen) was used. PCR was per-formed using MyCycler Thermal Cycler (Bio-Rad) in a total reaction mix-ture of 20 l containing 50 ng of cDNA, 1 HotStar TaqPCR buffer, and400 nM probe/primers mixture. After denaturation at 95C for 15 min, 55cycles were performed at 95C for 15 s, followed by 60C for 1 min.Comparative cycle threshold method with PCR efficiency correction, alsoknown as Pfaffls method, was used for quantification, as previously de-scribed (48). The expression levels of the target genes were adjusted to theexpression levels of the housekeeping genes PBGD and -actin.

    Cytokine determination

    Cytokines were measured using two different methods. The human cyto-kines IL-1, IL-2, IL-4, IL-5, IL-6, IL-7, IL-8, IL-10, IL-12p70, IL-13,

    IFN-, GM-CSF, and TNF- were simultaneously quantified in duplicateusing the human cytokine multiplex kit (Linco Research). Supernatantsobtained from day 7 (unstimulated DCs) or day 9 (DCs treated with mat-uration stimuli for 48 h) cultures were aliquoted and stored at 80C untilused. Sera from the same patients were also stored. Cytokine concentra-tions were determined following manufacturers instructions. In brief, stan-dards and samples were added into the appropriate wells. Mixed beadswere added to each well and the plate was incubated with agitation for 1 hat room temperature. Plate was washed and a detection Ab mixture wasadded to each well and incubated for 30 min at room temperature. Strepta-vidin-PE was added to each well containing the detection Ab mixture. Theplate was then incubated with agitation on a plate shaker for 30 min atroom temperature, washed, and sheath fluid was added for 5 min. Plate wasread on Luminex 100 (Luminex) and the concentration reported as pico-grams per milliliter.

    In addition, the BD CBA Human Inflammation kit (BD Biosciences)

    was used to quantitatively measure IL-8, IL-1, IL-6, IL-10, TNF-, andIL-12p70 protein levels in sera and supernatant samples, following man-ufacturers instructions. In brief, mixed capture beads were added to assaytubes. Human inflammation standard dilutions and test samples were addedto the assay tubes. Samples were incubated for 1.5 h at room temperatureand protected from direct exposure to light. Wash buffer was added andtubes were centrifuged at 200 g for 5 min. Supernatants were discardedleaving 100 l of liquid in each assay tube. Human inflammation PE de-tection reagent was added to the tubes and samples were incubated for 1.5 hat room temperature, washed, and analyzed by FACS using BD CytometricBead Array Software.

    T cell proliferation and determination of T cell activation

    T cells were isolated by negative selection using magnetic beads and in-structions provided by the manufacturer (pan T cell isolation kit; MiltenyiBiotec). Purity was 95%, as assessed by CD3 expression. T cell prolif-

    eration was analyzed using the intracellular fluorescent dye CFSE. Witheach cell division, the CFSE fluorescence intensity of the cells is reducedby half (4951). T cells from healthy controls were labeled with 2 MCFSE in RPMI 1640/10% FBS for 2 min at room temperature. After wash-ing to remove unbound fluorescent dye, 4 106 T cells were coculturedwith allogeneic DCs from SLE or healthy controls. Conditions includedunstimulated DCs or DCs stimulated with LPS and TNF- for 48 h, asdescribed above. Cells were cocultured at a T cell:DC ratio of 5:1 to 10:1.After 1 and 5 days, cells were harvested and stained with mouse anti-human CD3-allophycocyanin/Cy7, CD11c-PE, and CD25-CyChrome, thenfixed with 2% formaldehyde/PBS and analyzed by FACSCalibur, usingCellQuest software (BD Biosciences) and WinMDI. In additional experi-ments, expression of additional activation markers (CD69 and CD40L) wasmeasured on these T cells. Expression was measured 24 h and 5 days aftercocultures were started. Proliferation of T cells was analyzed by the log-arithmic reduction of CFSE staining of CD3-positive cells. The percentage

    of T cells that had undergone specific numbers of cell divisions (G1G5) orno cell divisions (G0) was calculated (52, 53). Controls included PHA-stimulated T cells and unstimulated T cells.

    Statistical analysis

    The difference between means was analyzed using paired t test or one-wayANOVA with post hoc analysis and Bonferroni correction. Spearman andPearsons correlation were used to assess correlation between differentvariables. Analyses were performed with SPSS version 11.5. A value ofp 0.05 was considered to be statistically significant.

    ResultsMyeloid lupus DCs display abnormal levels of differentiation

    and maturation markers and have abnormal responses tomaturation stimuli

    Confirming a previous study (54), no morphologic differences in

    the capacity of monocytes to differentiate into DCs were found

    between SLE and controls, using light and fluorescent microscopy

    (Fig. 1). After 57 days of culture, DCs from SLE patients and

    healthy controls were found to be increased in size and developed

    typical dendrites. To assess whether DCs from SLE individuals

    and controls differ in their differentiation and maturation poten-

    tials, monocyte-derived DCs from 31 individuals with SLE, 8 pa-

    tients with RA, and 20 healthy controls obtained at day 7 were

    stained with anti-human Abs to CD1a (to evaluate DC differenti-

    ation) (31) and CD83 (a marker of DC maturation) (55), and ex-

    pression of these markers was measured by FACS. As shown inFig. 2, AC, SLE patients, have increased numbers of cells ex-

    pressing the differentiation marker CD1a, and decreased numbers

    of cells expressing the maturation marker CD83 (p 0.05). No

    FIGURE 1. Monocyte-derived DCs from SLE patients display normal

    morphology. A, Images represent different magnifications of cells from one

    individual with SLE, after culture in X-vivo medium with IL-4 and GM-

    CSF for 7 days. Cells display the characteristic membrane and nuclear

    features of DCs. Arrows point at the cells that are displayed at higher

    magnification on the left panels. B, Fluorescent microscopy image of onecell from a patient with SLE that displays the classic morphology DCs, at

    day 7 in culture. The cell expresses both CD1a and CD83.

    5880 ABNORMAL PHENOTYPE AND FUNCTION OF LUPUS DCs

    p

    ,

    j

    g

    http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/
  • 7/30/2019 Sle Review

    5/13

    significant differences were observed in mean fluorescence inten-

    sity (data not shown). No significant differences were detected be-

    tween RA patients and controls in CD1a expression (Fig. 2A) or

    CD83 expression (Fig. 2B). The differences between SLE and con-

    trols were confirmed at the mRNA level by real-time RT-PCR

    (Fig. 2D). Indeed, lupus patients (but not RA patients) displayed

    significantly decreased mRNA levels of the monocyte marker

    CD14 and higher levels of CD1a mRNA at day 7, suggesting an

    acceleration of the differentiation from the monocyte stage to the

    myeloid DC stage (Fig. 2D). Baseline CD14 levels in monocytes

    did no differ between controls and lupus patients (data not shown),

    suggesting that the differences seen on day 7 DCs were the con-

    sequence of accelerated differentiation from the monocyte to the

    DC stage and not due to baseline down-regulation of CD14 on

    lupus monocytes. Interestingly, while CD83 protein and mRNA

    levels were significantly lower in lupus DCs at day 7 (Fig. 2, BD),

    FIGURE 2. Aberrant expression of differentiation and maturation markers in monocyte-derived DCs in SLE. Bar graph represents A, CD1a, and B, CD83

    percent expression, in 20 healthy controls, 8 RA patients, and 31 SLE patients. , p 0.05, results represent mean SEM of independent experiments.

    C, A representative density plot of CD83 and CD1a expression from one control and one SLE patient. D, Bar graphs represent CD1a, CD14, and CD83

    mRNA expression by real-time RT-PCR relative to housekeeping genes in day 7 DCs from 7 healthy controls, 22 SLE, and 7 RA patients. Results are

    reported as the fold change compared with healthy controls. , p 0.05, results represent mean SEM of independent experiments.

    5881The Journal of Immunology

    p

    ,

    j

    g

    http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/
  • 7/30/2019 Sle Review

    6/13

    mRNA levels of other specific maturation and differentiation markers

    (CD86, CD80, and HLA-DR) were significantly higher in SLE than

    in healthy controls in the absence of exogenous maturation stimuli

    (Fig. 3A). These findings were confirmed at the protein level (Fig. 3,

    B and C), both as numbers of cells expressing these markers (data not

    shown) and as mean fluorescent intensity in each cell. When com-

    pared with healthy controls, there were no differences in the expres-

    sion of maturation markers in RA patients (Fig. 3). These results in-

    dicate that differentiation and maturation are enhanced in monocyte-

    derived DCs from SLE patients, but that the maturation marker CD83

    is selectively down-regulated in lupus DCs.

    We considered the possibility that an in vivo serum effect could

    induce early responses in monocytes that would lead/promote the

    observed differences in maturation. However, when we treated lu-

    pus and healthy control monocytes with autologous sera, no dif-

    ferences were found between control and SLE sera on the induc-

    tion of expression of specific maturation markers (CD80: 11.5

    4.6% for untreated control DCs; 10.5 4.1% for control DCs

    treated with autologous sera; 32.6 11.2% for untreated lupus

    DCs and 34.2 12% for lupus DCs treated with autologous sera.

    CD83: 28.2 11.1% for untreated control DCs; 5.35 0.8% for

    control DCs treated with sera; 14.5 2% for untreated lupus DCs

    and 2 0.1 for lupus DCs treated with autologous serum, p NS

    when comparing lupus vs control sera; results represent mean

    SEM of three SLE patients and four healthy controls). These re-

    sults suggest that the differences found on maturation markers

    were intrinsic to lupus monocyte derived-DCs, rather than an ex-

    ogenous effect secondary to a serum factor or to serum withdrawal.

    Furthermore, when we cultured lupus monocytes in GM-CSF and

    IL-4 in the presence or absence of IFN-, there were no significant

    differences in the expression of CD83 between IFN-treated or un-

    treated cells, suggesting that adding this cytokine did not alter the

    phenotypic abnormalities seen in SLE DCs (CD83: 39 10% in

    IL-4 GM-CSF; 30 5% in IL-4 GM-CSF 100 U of

    IFN-; 30 4.4% in IL-4 GM-CSF 1000 U IFN-; results

    represent mean SEM of eight independent experiments, p 0.05).

    Similarly, when lupus monocytes were cultured in GM-CSF without

    IL-4 but in the presence of IFN-, this combination of cytokines did

    not result in changes in CD83 expression (10 4% in GM-CSF

    alone; 15 5% in GM-CSF 100 U of IFN-; 19 5% in GM-

    CSF 1000 U of IFN-; results represent mean SEM of eight

    independent experiments; p 0.05 between all conditions).To exclude the possibility that medications could account for the

    differences in expression of differentiation and maturation markers

    between controls and SLE patients, monocytes were treated with

    graded concentrations of drugs commonly used in the management of

    SLE, including hydrocortisone (for steroids), indomethacin (for non-

    steroidal anti-inflammatory drugs), chloroquine (for antimalarials),

    6-MP (for azathioprine), and MMF, while these cells were being dif-

    ferentiated into DCs in vitro. At doses equivalent to the ones used to

    treat SLE patients, we did not find any significant changes in the

    expression of differentiation and maturation markers induced by these

    drugs (Table II). Further, there were no significant correlations be-

    tween the use of specific immunosuppressive drugs by SLE patients

    and the phenotypic differences found in SLE DCs (Table III).Abnormal differentiation and maturation correlated with specific

    lupus clinical and serologic manifestations. Indeed, decreased

    CD83 expression in DCs correlated significantly with current or

    previous evidence of lupus nephritis, and with levels of anti-

    dsDNA Abs. Increased CD1a expression in lupus DCs correlated

    with decreased levels of complement (defined as C3 levels 83

    mg/dl and/or C4 levels 13 mg/dl) (Fig. 4B). These correlations

    were seen both at the protein (Fig. 4) and mRNA level (p 0.05

    for same variables). Increased levels of CD80 at the protein level

    correlated with hemologic manifestations of SLE (leucopenia and

    immune-mediated thrombocytopenia) (r 0.49, p 0.02) and

    levels of anti-dsDNA Abs (r 0.46, p 0.03), and negatively

    correlated with skin manifestations of SLE (r 0.59, p 0.004). Increased levels of CD86 at the protein level strongly cor-

    related with disease activity (SLEDAI) (r 0.74, p 0.001) and

    with previous or current lupus nephritis (r 0.45, p 0.04). The

    correlations between CD80 and CD86 with these specific clinical

    variables were also confirmed at the mRNA level (data not shown).

    To establish whether DCs from individuals with SLE normally

    up-regulate maturation markers in response to maturation stimuli,

    we proceeded to treat day 7 monocyte-derived DCs with LPS

    and/or TNF- for 48 h. As show in Fig. 5C, lupus DCs signifi-

    cantly up-regulate mRNA of the maturation markers CD86, CD80,

    and CD83. However, when compared with healthy controls, the

    degree of up-regulation for CD80 and CD86 was blunted in the

    lupus group both at the protein and mRNA level ( p 0.05) (Fig.

    5), using either LPS and/or TNF- as maturation stimuli. These

    experiments suggest that lupus DCs have the capacity to respond

    FIGURE 3. Increased expression of maturation markers in lupus DCs.

    A, Bar graphs represent mRNA expression of different maturation markers

    relative to housekeeping genes by real-time RT-PCR in day 7 DCs from 7

    healthy controls, 22 SLE patients, and 7 RA patients. Results are reported

    as the fold change compared with healthy controls. , p 0.05, results

    represent mean SEM of independent experiments. B, Bar graphs compare

    CD86 mean fluorescent intensity in day 7 DCs from 8 healthy controls, 5 RA

    patients, and 24 SLE patients. Representative histogram displays expression of

    CD86 on day 7 myeloid DCs from one healthy control, one RA patient, and

    one SLE patient. C, Bar graphs compare CD80 mean fluorescent intensity in

    day 7 DCs from 8 healthy controls, 5 RA patients, and 24 SLE patients.

    Representative histogram displays expression of CD80 on day 7 myeloid DCsfrom 1 healthy control, 1 RA patient, and 1 SLE patient. , p 0.05, results

    represent mean SEM of independent experiments.

    5882 ABNORMAL PHENOTYPE AND FUNCTION OF LUPUS DCs

    p

    ,

    j

    g

    http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/
  • 7/30/2019 Sle Review

    7/13

    to maturation stimuli, as shown by up-regulation of maturation

    markers; however, the level of up-regulation of these markers isdecreased when compared with healthy controls. When compared

    with healthy controls, there were no statistical differences in the

    capacity of RA DCs to up-regulate maturation markers after ex-

    posure to exogenous maturation stimuli (Fig. 5, A and B).

    To exclude the possibility that abnormal differentiation and mat-

    uration observed in SLE DCs were consequences of the in vitro

    conditions used, the expression of maturation and differentiation

    markers in myeloid DCs directly obtained from peripheral blood

    was examined. As shown in Fig. 6A, the myeloid DC population

    obtained directly from lupus blood is also characterized by higher

    numbers of CD1a cells, lower numbers of CD83 cells and

    higher expression of CD86 cells when compared with healthy

    controls, further confirming our in vitro findings. In addition, high

    levels of CD1a cells significantly correlated with levels of anti-

    dsDNA Abs (Fig. 6B) and with hypocomplementemia (p

    0.004). Furthermore, similar to what we found in DCs generated in

    vitro, there were no significant correlations between the use of

    specific immunosuppressive drugs and phenotypic abnormalities

    seen in lupus DCs isolated from peripheral blood (data not shown).

    Lupus DCs secrete higher levels of IL-8

    To evaluate whether SLE DCs show differential secretion of cy-

    tokines, we measured the secretion of a number of cytokines using

    cytometric bead array and a cytokine multiplex kit array. Control,

    RA, and SLE DCs secrete detectable amounts of IL-6, IL-7, IL-8,

    IL-10, IL-13, IFN-, and TNF-. The cytokine secreted at the

    highest levels was the proinflammatory cytokine IL-8, and lupusDCs secreted significantly higher levels of IL-8 than control DCs

    both before (5459.2 813.7 vs 2137.9 759.3 pg/ml, respec-

    tively, mean SEM, p 0.05) and after TNF- stimulation

    (8760.6 1048.6 vs 4817.2 689.4, respectively, mean SEM,

    p 0.05) (Fig. 7A). Levels of IL-8 secreted by lupus DCs corre-lated with expression of the maturation marker CD80 (r 0.56,

    p 0.01, Pearsons correlation), and patients with higher produc-

    tion of IL-8 had higher anti-dsDNA (Fig. 7B) and anti-cardiolipin

    levels in sera (r 0.53, p 0.007 and r 0.45, p 0.02,

    respectively, Pearsons correlation). When compared with healthy

    controls, there were no significant increases in IL-8 secretion by

    DCs from RA patients (Fig. 7B). No significant differences be-

    tween control, SLE, and RA patients were observed when the other

    cytokines were measured in DC supernatants (data not shown). Of

    interest, IL-8 was also the most abundant cytokine detected in

    plasma from SLE patients and serum levels were higher than in

    healthy controls, although no statistical significance was found

    (57.9 vs 36.3 pg/ml, P NS).

    Lupus DCs up-regulate T cell proliferation and activation

    Unstimulated and stimulated DCs from lupus patients induced a

    significant increase in allogeneic T cell proliferation when com-

    pared with DCs from healthy controls. As expected, unstimulated

    DCs from healthy controls were poor stimulators of allogeneic T

    cell proliferation and, after stimulation with maturation stimuli,

    their capacity to induce allogeneic T cell proliferation increased

    significantly. When cocultured with lupus DCs, allogeneic T cells

    showed a significantly higher percentage of proliferating cells than

    what was observed with control allogeneic DCs, and the effect was

    more pronounced in DCs treated with maturation stimuli (LPS and

    TNF-) (Fig. 8, A and B). This increase in proliferation required

    cell-cell interactions, as supernatants from lupus DCs did not in-duce similar increases in T cell proliferation (data not shown).

    Similarly, there was a statistically significant increase in the ex-

    pression of the activation marker CD40L in allogeneic T cells

    Table III. Correlation analysis of specific immunosuppressive/immunomodulator use and expression of differentiation and maturation markers inlupus DCsa

    Corticosteroids Antimalarials Azathioprine Mycophenolate

    CD1a p 0.24 (0.28) p 0.50 (0.16) p 0.63 (0.121) p 0.46 (0.15)CD83 p 0.77 (0.07) p 0.12 (0.37) p 0.29 (0.262) p 0.16 (0.24)CD86 p 0.14 (0.31) p 0.99 (0.0002) p 0.71 (0.08) p 0.47 (0.17)CD80 p 0.65 (0.11) p 0.96 (0.009) p 0.47 (0.18) p 0.24 (0.27)

    IL-8 p 0.42 (0.20) p 0.30 (0.26) p 0.30 (0.18) p 0.52 (0.16)

    a Results represent p value (r ), using Spearman correlation. Negative numbers indicate a negative correlation. All p values were 0.05.

    Table II. Expression of maturation and differentiation markers on day 7 monocyte-derived DCs, after in vitro drug treatmenta

    CD1a CD83 CD86 CD80

    Untreated 18.08 6.5 24.86 0.27 34 1.67 89.2 0.4HC 0.01 M 19.08 1.76 39.9 9.6 36.7 0.07 84 0.3HC 0.1 M 24 5.01 27.1 4.2 28 10.47 82.5 0.5HC 1 M 31 9 26.06 6.7 26.5 3.2 83.2 26-MP 0.01 M 10.25 0.2 33.5 2.7 46.7 3.45 84.4 0.96-MP 0.1 M 13.3 0.25 27.2 4.2 36.6 0.075 82.2 0.8

    6-MP 1 M 14.8 0.9 35.4 4.9 47 3.5 84.1 3INDO 0.01 g/ml 10.6 5.6 34.4 0.08 33.5 1.7 83 9INDO 0.1 g/ml 14.6 0.52 35.2 1 35 16.22 77.8 5INDO 1 g/ml 15.5 1.4 35.6 6.5 32.3 16.2 77 6MMF 0.04 g/ml 14 8.5 30.9 8.3 51 0.5 92 5MMF 0.4 g/ml 10.7 5.7 32 11.5 51 11 93 2.8MMF 4 g/ml 18 3.5 37 18.4 41 2.05 94 3HCQ 0.02 g/ml 13.5 7.1 28 10.5 51 .3 89 2HCQ 0.2 g/ml 6.9 2.85 31 8.3 57 11.2 93 3HCQ 2 g/ml 3.44 1.4 28.7 11.6 51 6 91 1.1

    a HC, Hydrocortisone; 6-MP, 6-mercaptopurine; INDO, Indomethacin; MMF, mycophenolate mofetil; HCQ, hydroxychloroquine. Vehicle results did not differ from resultsfrom untreated cells. Values represent mean SEM of three independent experiments and all values are p 0.05 when compared to untreated cells.

    5883The Journal of Immunology

    p

    ,

    j

    g

    http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/
  • 7/30/2019 Sle Review

    8/13

    when they were cocultured for 24 h with either unstimulated or

    stimulated lupus DCs, as compared with DCs from healthy con-

    trols (Fig. 8C). An increase in the percentage of cells that coex-

    press CD25 and CD40L in allogeneic T cells was also observed

    after these cells were cocultured with either unstimulated or stim-

    ulated lupus DCs for 5 days, as compared with unstimulated or

    stimulated DCs from healthy controls (percent of CD25

    CD40L

    T cells: 2.4 1% with unstimulated control DCs were added;

    5.8 2.1% when unstimulated lupus DCs were added; 4.8 1.2%

    when stimulated control DCs were added;. 10.8 3% when stim-

    ulated lupus DCs were added; p 0.05 between control and lupus

    cells for both conditions; results represent mean SEM of inde-

    pendent experiments using five controls and 11 SLE patients). No

    significant differences were seen for the early T cell activation

    marker CD69 between control and lupus DCs (data not shown). As

    expected, DCs treated with maturation stimuli were more effective

    at inducing allogeneic T cell activation (Fig. 8).

    Discussion

    This study establishes that myeloid DCs from individuals with SLEdisplay distinct phenotypic and functional differences characteristic of

    proinflammatory DCs and promote abnormal T cell responses. These

    phenotypic changes are characterized by accelerated differentiation

    from the monocyte to the myeloid DC stage (as measured by in-

    creased expression of the DC differentiation marker CD1a), up-regu-

    lation of different costimulatory molecules which are involved in T

    cell priming, even in the absence of exogenous maturation stimuli;

    and increased production of the proinflammatory cytokine IL-8. In

    addition, myeloid DCs from SLE patients display a selective down-

    regulation of the maturation marker CD83. These abnormalities were

    seen not only on in vitro-derived DCs, but also in DCs obtained from

    peripheral blood, indicating that this phenomenon is not the conse-

    quence of added cytokines in vitro or a serum withdrawal effect but

    likely represent true abnormalities on the differentiation/maturation

    pathways of myeloid DCs in vivo.

    These phenotypic differences correlate with specific clinical and

    serologic manifestations of the disease. Because the population

    studied was fairly well-controlled, with an SLEDAI of 4.5 3

    (mean SEM), the results of this study suggest that the abnor-

    malities observed in SLE DCs are not merely the result of active

    disease but might have pathogenic significance. Further, these phe-

    notypic differences in lupus DCs likely have functional relevance,as both unstimulated and stimulated myeloid DCs from lupus pa-

    tients induce significant increases in allogeneic T cell proliferation

    and activation, when compared with healthy controls. Immature

    DCs are known to have a low T cell activation potential (20) and

    this promotion of T cell proliferation and activation is likely the

    consequence of an increased mature phenotype. Accelerated dif-

    ferentiation from the monocyte (or potentially other myeloid pre-

    cursors) to the DC stage, and up-regulation of maturation markers

    could promote and enhance the capabilities of lupus DCs to prime

    and activate T cells in the spleen and other lymphoid organs, fur-

    ther contributing to the T cell hyperresponsiveness and enhanced

    activation described in SLE. Supporting this idea, a recent study

    reported up-regulation of CD11b

    CD11c

    DCs in the thymus andspleen in aged BWF1 lupus-prone mice (56), suggesting acceler-

    ated migration to these organs. Studies in humans have also shown

    that myeloid and plasmacytoid DCs are markedly decreased in

    SLE blood and it has been speculated, although not proven, that

    this might reflect an accelerated migration of these cells from the

    blood into tissues (22, 57). Certainly, the up-regulation of CD86

    and other maturation markers in DCs would support the possibility

    that a mature DC phenotype would be associated to increased

    migration to lymphoid organs of SLE DCs and stimulation and

    priming of T cells in these organs. DCs can induce either T cell

    tolerance or strong innate and adaptive immunity to specific Ag. In

    general, tolerance is initiated when DCs are immature, whereas the

    initiation of immunity requires an effective DC maturation signal.

    Therefore, accelerated DC maturation in SLE in the absence of

    extrinsic danger signals suggests that these cells can become very

    FIGURE 4. Aberrant differentia-

    tion and maturation in SLE correlatewith clinical and serologic features of

    lupus. A, Correlation of a low percent

    of CD83-positive day 7 DCs, with

    past or current lupus nephritis and

    with levels of anti-dsDNA Abs. B,

    Correlation of an increased percent of

    CD1a-positive day 7 DCs with serum

    complement levels; , p 0.05,

    mean SEM of independent exper-

    iments. Similar correlations were

    seen at the mRNA level.

    5884 ABNORMAL PHENOTYPE AND FUNCTION OF LUPUS DCs

    p

    ,

    j

    g

    http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/
  • 7/30/2019 Sle Review

    9/13

    efficient autoantigen-presenting cells and drive autoimmune re-

    sponses. In addition, the role of costimulatory molecules is well

    documented in murine models of lupus, including the observation

    that treatment with a combination of blocking mAbs to CD80 and

    CD86, before the onset of murine lupus, significantly improves

    survival and severity of the disease (58, 59).

    Given that mature myeloid DCs are able to break tolerance and

    induce lupus autoantibodies in normal hosts (60), the increase in DC

    maturation in SLE suggests that these abnormalities might be very

    relevant in the induction and perpetuation of autoimmunity in SLE.

    Increased secretion of IL-8 by lupus DCs could potentially con-

    tribute to tissue damage, as this cytokine has been proposed to play

    an important role in the development of lupus nephritis (6164)

    and has been found to be elevated in lupus patients with CNS in-

    volvement (65). IL-8 is produced by numerous cell types, includingmonocytes/macrophages and DCs (64, 66, 67). IL-8 is mainly active

    on neutrophils, promoting their recruitment and also their strong ac-

    tivation which triggers the leukotriene pathway, induces the release of

    their granular content, elastase and lactoferrin and increases their ad-

    herence to endothelial cells (6871). In fact, migration of neutrophils

    is influenced by DCs primarily by IL-8 (64). IL-8 is also a chemoat-

    tractant for other cell types including T cells (72). In addition, we

    found a significant association between IL-8 secretion and serum lev-

    els of anti-dsDNA. Anti-dsDNA can enhance the release of proin-

    flammatory cytokines, including IL-8 and TNF- from mononuclear

    cells to augment inflammatory reactions and can polarize the immune

    reaction toward the Th2 pathway (7274). Therefore, an increase in

    the production of this cytokine by DCs might enhance the ability torecruit cells in the glomerulus and enhance neutrophil adhesion to

    vascular endothelium which in turn may contribute to renal and vas-

    cular damage. Interestingly, a recent study has shown that the IL-8

    gene and its receptor CXCR-2 are up-regulated in PBMCs from SLE

    patients using microarrays (75).

    The functional relevance of decreased levels of CD83 in SLE is

    unclear at this point. CD83 is a cell surface membrane glycopro-

    tein whose surface expression is largely restricted to DCs (55). The

    precise functions of this molecule remain unknown (76, 77), but

    CD83 may serve important roles during intercellular interactions

    (7779), as membrane-bound CD83 increases the stimulatory ca-

    pacity of DCs (79). Further, previous studies suggest that CD83

    mediates adhesion to monocytes and CD8 T cells (80). CD83-Ig

    enhances T cell proliferation and increases the proportion of CD8

    T cells (80), and engagement of CD83 delivers a significant signal

    FIGURE 5. Responses of monocyte-derived DCs to maturation stimuli

    are abnormal in lupus DCs. A, Bar graphs display mean fluorescence in-

    tensity of CD86 in DCs from 10 healthy controls, 5 RA patients, and 20

    SLE patients before and after stimulation with TNF-. B, Representative

    histograms of CD86 expression on DCs from 1 healthy control, 1 patient

    with SLE, and 1 patient with RA before and after stimulation with LPS and

    TNF-. C, Bar graphs display mRNA expression by real-time RT-PCR of

    differentiation and maturation markers normalized to housekeeping genes

    in DCs, after stimulation with LPS and TNF- in 6 healthy controls and 17

    SLE patients. Results are reported as the fold change when compared with

    unstimulated day 7 DCs. , p 0.05 when compared before and after

    stimulation; results are presented as mean SEM.

    FIGURE 6. Aberrant expression of differentiation and maturation mark-

    ers in lupus myeloid DCs obtained from peripheral blood. A, Bar graphs

    display expression of DC differentiation and maturation markers in freshly

    isolated CD11c cells from peripheral blood, in 11 healthy controls and 24

    SLE patients. B, Bar graphs display correlation of anti-dsDNA Abs with

    CD1aDCs in peripheral blood. , p 0.05, results represent the mean

    SEM of independent experiments.

    5885The Journal of Immunology

    p

    ,

    j

    g

    http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/
  • 7/30/2019 Sle Review

    10/13

    specifically supporting the expansion, survival and function of

    newly primed naive CD8 T cells (81). CD8 T cells in lupus-

    prone mice are impaired in expansion, acquisition of memory, se-

    cretion of cytokine, and suppression of autoimmunity (8, 82) and

    because CD83 appears to have a role in CD8 function, it is pos-

    sible that down-regulation of the former could contribute to ab-

    normal CD8 function in SLE. In addition, CD4 T cells that

    develop in a CD83 mutant animal fail to respond normally fol-lowing allogeneic stimulation (83), at least in part due to an altered

    cytokine expression pattern characterized by an increased produc-

    tion of IL-4 and IL-10 and diminished IL-2 production, findings

    typically seen in SLE. Thus, absence or decrease of CD83 in SLE

    DCs may result in the generation of T cells with an altered acti-

    vation and cytokine profile. Future studies will address these pos-

    sibilities. The factors inducing down-regulation of CD83 in lupus

    DCs remain to be determined. Although we detected that autolo-

    gous serum down-regulates the percentage of DCs that express

    CD83, this phenomenon was observed for both lupus and control

    serum, and therefore cannot explain the differences in expression

    of this molecule observed between the two groups. Similarly, treat-

    ment with IFN- did not alter CD83 expression.

    DCs from SLE patients responded to extrinsic maturation stimuli

    by up-regulating the expression of maturation markers, further in-

    creasing IL-8 secretion and enhancing T cell proliferative and activa-

    tion responses. However, the up-regulation of some of the membrane-

    bound maturation markers was blunted when compared with the

    degree of up-regulation seen in healthy controls. It is possible that the

    preactivation status of lupus DCs, as confirmed by the overexpression

    of maturation markers even before maturation stimuli, makes the lu-

    pus cells more refractory to further up-regulation of cell surface mat-

    uration markers. Lupus DCs treated with maturation stimuli did be-

    come more efficient at inducing allogeneic T cell proliferation, T cell

    activation and cytokine secretion, than untreated lupus DCs.

    Although we cannot entirely exclude the possibility that immuno-

    suppressive drugs could play a role in the phenotypic and functional

    differences of lupus myeloid DCs, correlation analysis did not show

    an association between expression of specific differentiation and mat-

    uration markers and secreted cytokines with different medications

    used by SLE patients. Further, when we treated monocytes with drugs

    commonly used to treat SLE and differentiated them into DCs, we

    found no differences in the maturation and differentiation patterns

    seen in untreated or treated cells. Also, while RA patients are also

    exposed to immunosuppressive medications, DCs from patients with

    this disease did not show any significant differences in the expression

    of maturation and differentiation markers and cytokine profile when

    compared with healthy controls. These observations suggest that ourfindings are not related to the use of medications in these patients. In

    addition, an effect of corticosteroid treatment on DC function can

    reasonably be excluded, as these agents have a short biological half-

    life of 1236 h (84) and therefore can only have a marginal effect on

    maturation of DCs after a 7-day culture. Further supporting that our

    results are not related to medication use, a recent report in a novel

    lupus-prone mouse strain, B6.Sle3 (not subject to any type of immu-

    nosuppressive treatment), indicates that its DCs are hyperstimulatory,

    more mature and proinflammatory, overexpress CD80 and CD86

    among other costimulatory molecules, and induce T cell hyperactivity

    (85), similar to our findings in human lupus DCs.

    Our results on the differentiation and maturation status of my-

    eloid derived DCs differ from the ones previously described byKoller et al. (54), where no significant increases in the expression

    of maturation markers in lupus DCs and a decrease in T cell pro-

    liferation by lupus DCs were found. We believe that these discrep-

    ancies might be secondary to differences in methodology. The

    number of patients studied by Koller et al. (54) was significantly

    smaller and disease activity also appeared to be lower, although a

    different scoring system was used. That group used positive selec-

    tion to isolate monocytes, which could contribute to activation of

    these cells, as has been reported for other PBMCs (86). In addition,

    their cells were cultured for longer periods of time and under con-

    ditions that were not serum-free. It has been demonstrated that

    serum contains growth factors that could affect DC development,

    differentiation and maturation in vitro (87, 88) and that serum-freeconditions like the ones used in our study lead to more optimal DC

    harvest that non-serum free conditions (89). Furthermore, Koller et

    al. (54) used irradiated DCs for their mixed-lymphocyte reaction

    studies, while we used nonirradiated DCs. Irradiated DCs can un-

    dergo apoptosis (90) and posttranslational modifications of pro-

    teins during apoptosis can potentially modify T cell activation and

    proliferation (90). Our results do confirm a previous observation

    that CD83 expression on lupus DCs is not up-regulated after mat-

    uration stimuli (91), are supported by similar ex vivo findings in

    DCs isolated from peripheral blood and, as mentioned, are remi-

    niscent of what has been reported in lupus animal models (85).

    Our observations that DCs from RA patients do not display the

    phenotypic abnormalities seen in SLE are in consensus what has

    been previously been reported in the RA literature in monocyte-

    derived DCs (92, 93). Our observations may not necessarily indicate

    FIGURE 7. Increased IL-8 secretion by myeloid DCs from SLE pa-

    tients. A, Results represent the mean concentration in supernatants (pico-

    grams per milliliter) SEM in DC cultures from 12 healthy controls and

    27 SLE patients. Concentrations were adjusted according to initial mono-

    cyte number; , p 0.05 when comparing controls vs SLE in each culture

    condition. B, Results represent the mean concentration in supernatants (pi-

    cograms per milliliter) SEM in DC cultures from 7 healthy controls, 5

    RA patients, 13 SLE patients with negative anti-dsDNA Abs, and 9 SLEpatients with elevated anti-dsDNA Abs. The variability in measurements

    between A and B is due to different methodology (CBA for A, Luminex

    array for B) and different samples measured.

    5886 ABNORMAL PHENOTYPE AND FUNCTION OF LUPUS DCs

    p

    ,

    j

    g

    http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/
  • 7/30/2019 Sle Review

    11/13

    that all RA DCs have normal phenotype and function. Indeed,

    monocyte-derived DCs might not reflect changes occurring in the

    inflamed synovium and there is evidence that DCs in the joint of

    RA patients show a distinct phenotype, with differential expression

    of specific TLRs (94). DCs are abundant both in synovial tissueand in synovial fluid of RA patients (95) and it has been proposed

    that RA synovitis may be a delayed-type hypersensitivity reaction

    generated by the interaction of synovial DCs and T cells (96).

    Furthermore, the cytokine profile in RA and SLE is known to be

    distinctly different (35, 93, 97) and there is evidence that the mech-

    anisms leading to autoimmunity in SLE and RA are probably quite

    distinct (98, 99). Therefore, the lack of hyperactivated DCs in RA

    patients does not imply that the abnormal DC phenotype observed

    in SLE is not important in maintaining autoimmunity, and this

    statement is supported by findings in lupus animal models (56, 85).

    Immune complexes consisting of DNA and anti-dsDNA Abs

    isolated from the sera of patients with SLE can induce plasmacy-

    toid DCs to produce high levels of IFN- (100). Increased IFN-

    production in SLE causes increased monocyte differentiation into

    DCs (22). These cells are able to capture apoptotic cells and

    present their Ags to autologous T cells and induce potent MLRs

    (22). We propose that, in addition to the IFN-, there are endog-

    enous abnormalities in myeloid DC differentiation and maturation,

    as a serum effect and exogenous IFN- could not account for, or

    abrogate, the differences in phenotype and function seen in lupusmonocyte-derived DCs. Purified nucleosomes directly induce in

    vitro DC maturation of mouse bone marrow-derived DC, human

    monocyte-derived DC and purified human myeloid DCs as ob-

    served by stimulation of allogenic cells in MLR, IL-8 secretion,

    and CD86 up-regulation (101), findings similar to the ones we

    report in this study. Further, nucleosomes complexed with antinu-

    cleosome Abs can activate DCs (102). Therefore, autoantigens and

    immune complexes could play an important role in vivo by induc-

    ing accelerated differentiation and maturation of lupus DCs.

    Taken together, our data suggest that monocyte-derived DC dif-

    ferentiation and maturation are altered in SLE and contribute to

    enhanced T cell proliferation and activation and to an increase in

    the secretion of proinflammatory cytokines. We hypothesize that

    these events could help to initiate and maintain the autoimmune

    response in lupus.

    FIGURE 8. Lupus DCs promote increased al-

    logeneic T cell proliferation and activation. A,

    Bar graph represents percentage of allogeneic T

    cells in phases G0-G5, after coculture for 5 days

    with either unstimulated or stimulated (LPS

    TNF-) DCs from 11 healthy controls and 19

    SLE patients. Proliferation of untreated T cells

    and PHA-treated T cells treated is also shown.

    Results represent the mean SEM of indepen-

    dent experiments. , p 0.05 when comparing

    proliferation of T cells adding SLE DCs with

    healthy control DCs and with untreated T cells.

    , p 0.05 when comparing proliferation of T

    cells stimulated with healthy control DCs vs un-

    treated T cells. Both control and SLE-stimulated

    DCs induced higher T cell proliferation rates

    when compared with unstimulated healthy con-

    trol or lupus DCs (p 0.02 for G0 in SLE; p

    0.02 for G2

    in control; p 0.04 for G2 in SLE

    and p 0.04 for G3 in control). B, Representa-tive dot plots display CFSE and CD25 expres-

    sion in allogeneic T cells after exposure to stim-

    ulated DCs from 1 SLE patient or DCs from 1

    healthy control. C, Bar graph represents percent-

    age of CD40L allogeneic T cells after 24 h co-

    culture with unstimulated or stimulated (LPS

    TNF-) DCs from 4 healthy controls and 7 SLE

    patients. Untreated T cells and PHA-treated T

    cells treated are also shown. Results represent

    the mean SEM of independent experiments. ,

    p 0.01 when comparing unstimulated SLE vs

    unstimulated healthy control DCs and p 0.03

    when comparing stimulated lupus DCs vs stim-

    ulated control DCs. There was also a statistically

    significant difference (p 0.04) in up-regulationof CD40L between untreated and stimulated

    DCs for both control and SLE patients.

    5887The Journal of Immunology

    p

    ,

    j

    g

    http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/
  • 7/30/2019 Sle Review

    12/13

    AcknowledgmentsWe thank Drs. David Fox, Bruce Richardson, and Michael Denny for crit-

    ical review of the manuscript; Joel Joshua for help in patient recruitment,

    and Berlex for providing human GM-CSF.

    DisclosuresThe authors have no financial conflict of interest.

    References1. Tsokos, G. C., H. Wong, E. Enyedi, and M. Nambiar. 2000. Immune cell sig-naling in lupus. Curr. Opin. Rheumatol. 12: 355363.

    2. Kammer, G., A. Perl, B. Richardson, and G. Tsokos. 2002. Abnormal T cellsignal transduction in systemic lupus erythematosus. Arthritis Rheum. 46:11391154.

    3. Tsokos, G. C., J. Mitchell, and Y. Juang. 2003. T cell abnormalities in human andmouse lupus: intrinsic and extrinsic. Curr. Opin. Rheumatol. 15: 542547.

    4. Busser, B. W., B. Adair, J. Erikson, and T. Laufer. 2003. Activation of diverserepertoires of autoreactive T cells enhances the loss of anti-dsDNA B cell toler-ance. J. Clin. Invest. 112: 13611371.

    5. Riemekasten, G., D. Langnickel, F. Ebling, G. Karpouzas, J. Kalsi, G. Herberth,B. Tsao, P. Henklein, S. Langer, G. Burmester, et al. 2003. Identification andcharacterization of SmD183119-reactive T cells that provide T cell help forpathogenic anti-double-stranded DNA antibodies. Arthritis Rheum 48: 475485.

    6. Kaplan, M. J., Q. Lu, A. Wu, J. Attwood, and B. Richardson. 2004. Demethyl-ation of promoter regulatory elements contributes to perforin overexpression inCD4 lupus T cells. J. Immunol. 172: 36523661.

    7. Tsai, H. F., J. Lai, A. Chou, T. Wang, C. Wu, and P. Hsu. 2004. Induction ofcostimulation of human CD4 T cells by tumor necrosis factor-related apoptosis-inducing ligand: possible role in T cell activation in systemic lupus erythemato-sus. Arthritis Rheum 50: 629 639.

    8. Karpouzas, G. A., A. La Cava, F. M. Ebling, R. R. Singh, and B. H. Hahn. 2004.Differences between CD8 T cells in lupus-prone (NZB NZW)F

    1mice and

    healthy (BALB/c NZW)F1

    mice may influence autoimmunity in the lupusmodel. Eur. J. Immunol. 34: 24892499.

    9. Wenzel, J., S. Henze, S. Brahler, T. Bieber, and T. Tuting. 2005. The expressionof human leukocyte antigen-DR and CD25 on circulating T cells in cutaneouslupus erythematosus and correlation with disease activity. Exp. Dermatol. 14:454459.

    10. Zielinski, C. E., S. Jacob, F. Bouzahzah, B. Ehrlich, and J. Craft. 2005. NaiveCD4 T cells from lupus-prone Fas-intact MRL mice display TCR-mediatedhyperproliferation due to intrinsic threshold defects in activation. J. Immunol.174: 51005109.

    11. Kaplan, M. J., E. Lewis, E. Shelden, E. Somers, R. Pavlic, W. McCune, andB. Richardson. 2002. The apoptotic ligands TRAIL, TWEAK, and Fas ligandmediate monocyte death induced by autologous lupus T cells. J. Immunol. 169:60206029.

    12. Calvani, N., H. Richards, M. Tucci, G. Pannarale, and F. Silvestris. 2004. Up-regulation of IL-18 and predominance of a Th1 immune response is a hallmarkof lupus nephritis. Clin. Exp. Immunol. 138: 171178.

    13. Gomez, D., P. Correa, L. Gomez, J. Cadena, J. Molina, and J. Anaya. 2004.Th1/Th2 cytokines in patients with systemic lupus erythematosus: is tumor ne-crosis factor protective? Semin. Arthritis Rheum. 33: 404 413.

    14. Zeng, D., Y. Liu, S. Sidobre, M. Kronenberg, and S. Strober. 2003. Activation ofnatural killer T cells in NZB/W mice induces Th1-type immune responses ex-acerbating lupus. J. Clin. Invest. 112: 12111222.

    15. Singh, R. 2003. IL-4 and many roads to lupus like autoimmunity. Clin. Immunol.108: 7379.

    16. Nakajima, A., S. Hirose, H. Yagita, and K. Okumura. 1997. Roles of IL-4 andIL-12 in the development of lupus in NZB/W F

    1mice. J. Immunol. 158:

    14661472.17. Tyrrell-Price, J., P. Lydyard, and D. Isenberg. 2001. The effect of interleukin-10

    and of interleukin-12 on the in vitro production of anti-double-stranded DNAantibodies from patients with systemic lupus erythematosus. Clin. Exp. Immunol.

    124: 118125.18. Llorente, L., W. Zou, Y. Levy, Y. Richaud-Patin, J. Wijdenes, J. Alcocer-Varela,

    B. Morel-Fourrier, J. Brouet, D. Alarcon-Segovia, P. Galanaud, and D. Emilie.1995. Role of interleukin 10 in the B lymphocyte hyperactivity and autoantibodyproduction of human systemic lupus erythematosus. J. Exp. Med. 181: 839844.

    19. Palucka, A. K., J. Banchereau, P. Blanco, and V. Pascual. 2002. The interplay ofdendritic cell subsets in systemic lupus erythematosus. 80: 484488.

    20. Banchereau, J., and R. M. Steinman. 1998. Dendritic cells and the control ofimmunity. Nature 392: 245252.

    21. Kono, D. H., R. Baccala, and A. Theofilopoulos. 2003. Inhibition of lupus bygenetic alteration of the interferon-/ receptor. Autoimmunity 36: 503510.

    22. Blanco, P., A. Palucka, M. Gill, V. Pascual, and J. Banchereau. 2001. Inductionof dendritic cell differentiation by IFN- in systemic lupus erythematosus. Sci-ence 294: 15401543.

    23. Stoll, S., J. Delon, T. M. Brotz, and R. N. Germain. 2002. Dynamic imaging ofT cell-dendritic cell interactions in lymph nodes. Science 296: 18731876.

    24. Mailliard, R. B., S. Egawa, Q. Cai, A. Kalinska, S. N. Bykovskaya, M. T. Lotze,M. L. Kapsenberg, W. J. Storkus, and P. Kalinski. 2002. Complementary den-

    dritic cell-activating function of CD8

    and CD4

    T cells: helper role of CD8

    T cells in the development of T helper type 1 responses. J. Exp. Med. 195:473483.

    25. Angelini, G., S. Gardella, M. Ardy, M. R. Ciriolo, G. Filomeni, G. Di Trapani,F. Clarke, R. Sitia, and A. Rubartelli. 2002. Antigen-presenting dendritic cellsprovide the reducing extracellular microenvironment required for T lymphocyteactivation. Proc. Natl. Acad. Sci. USA 99: 14911496.

    26. Cella, M., D. Scheidegger, K. Palmer-Lehmann, P. Lane, A. Lanzavecchia, andG. Alber. 1996. Ligation of CD40 on dendritic cells triggers production of highlevels of interleukin-12 and enhances T cell stimulatory capacity: T-T help viaAPC activation. J. Exp. Med. 184: 747752.

    27. Daniel, P. T., C. Scholz, J. Westermann, B. Dorken, and A. Pezzutto. 1998.Dendritic cells prevent CD95 mediated T lymphocyte death through costimula-tory signals. Adv. Exp. Med. Biol. 451: 173177.

    28. Ingulli, E., D. R. Ulman, M. M. Lucido, and M. K. Jenkins. 2002. In situ analysisreveals physical interactions between CD11b dendritic cells and antigen-specificCD4 T cells after subcutaneous injection of antigen. J. Immunol. 169: 22472252.

    29. Maldonado-Lopez, R., T. De Smedt, P. Michel, J. Godfroid, B. Pajak,C. Heirman, K. Thielemans, O. Leo, J. Urbain, and M. Moser. 1999. CD8 andCD8 subclasses of dendritic cells direct the development of distinct T helpercells in vivo. J. Exp. Med. 189: 587592.

    30. Sallusto, F., and A. Lanzavecchia. 2002. The instructive role of dendritic cells onT-cell responses. Arthritis Res. 4(Suppl. 3): S127S132.

    31. Brigl, M., and M. Brenner. 2004. CD1: antigen presentation and T cell function.Annu. Rev. Immunol. 22: 817890.

    32. Langenkamp, A., G. Casorati, C. Garavaglia, P. Dellabona, A. Lanzavecchia, andF. Sallusto. 2002. T cell priming by dendritic cells: thresholds for proliferation,differentiation and death and intraclonal functional diversification. Eur. J. Immu-nol. 32: 20462054.

    33. Penna, G., M. Vulcano, A. Roncari, F. Facchetti, S. Sozzani, and L. Adorini.2002. Differential chemokine production by myeloid and plasmacytoid dendriticcells. J. Immunol. 169: 66736676.

    34. Pulendran, B., J. L. Smith, G. Caspary, K. Brasel, D. Pettit, E. Maraskovsky, and

    C. R. Maliszewski. 1999. Distinct dendritic cell subsets differentially regulate theclass of immune response in vivo. Proc. Natl. Acad. Sci. USA 96: 10361041.

    35. Banchereau, J., V. Pascual, and A. Palucka. 2004. Autoimmunity through cyto-kine-induced dendritic cell activation. Immunity 20: 539550.

    36. Santiago-Raber, M. L., R. Baccala, K. Haraldsson, D. Choubey, T. Stewart,D. Kono, and A. Theofilopoulos. 2003. Type-I interferon receptor deficiencyreduces lupus-like disease in NZB mice. J. Exp. Med. 197: 777788.

    37. Tan, E. M., A. Cohen, J. Fries, A. Masi, D. McShane, N. Rothfield, J. Schaller,N. Talal, and R. Winchester. 1982. The 1982 revised criteria for the classificationof systemic lupus erythematosus. Arthritis Rheum. 25: 12711277.

    38. Arnett, F. C., S. Edworthy, D. Bloch, D. McShane, J. Fries, N. Cooper, L. Healey,S. Kaplan, M. Liang, H. Luthra, et al. 1988. The American Rheumatism Asso-ciation 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis

    Rheum. 31: 315324.

    39. Hochberg, M. C., for the Diagnostic and Therapeutic Criteria Committee of theAmerican College of Rheumatology. 1997. Updating the American College ofRheumatology revised criteria for the classification of systemic lupus erythem-atosus [letter]. Arthritis Rheum. 40: 1725.

    40. Bombardier, C., D. Gladman, M. Urowitz, D. Caron, and C. Chang. 1992. Der-ivation of the SLEDAI: a disease activity index for lupus patients. The Commit-tee on Prognosis Studies in SLE. Arthritis Rheum. 35: 630 640.

    41. Scandella, E., Y. Men, S. Gillessen, R. Forster, and M. Groettrup. 2002. Prosta-glandin E

    2is a key factor for CCR7 surface expression and migration of mono-

    cyte-derived dendritic cells. Blood 100: 13541361.

    42. Munster, T., J. P. Gibbs, D. Shen, B. A. Baethge, G. R. Botstein, J. Caldwell,F. Dietz, R. Ettlinger, H. E. Golden, H. Lindsley, et al. 2002. Hydroxychloro-quine concentration-response relationships in patients with rheumatoid arthritis.

    Arthritis Rheum. 46: 14601469.43. Ensom, M. H., N. Partovi, D. Decarie, A. P. Ignaszewski, G. J. Fradet, and

    R. D. Levy. 2003. Mycophenolate pharmacokinetics in early period followinglung or heart transplantation. Ann. Pharmacother. 37: 17611767.

    44. Doria, A., M. Cutolo, A. Ghirardello, S. Zampieri, F. Vescovi, A. Sulli,M. Giusti, A. Piccoli, P. Grella, and P. F. Gambari. 2002. Steroid hormones anddisease activity during pregnancy in systemic lupus erythematosus. Arthritis

    Rheum. 47: 202209.45. Iqbal, M. P., J. A. Baig, A. A. Ali, S. K. Niazi, N. Mehboobali, and

    M. A. Hussain. 1998. The effects of non-steroidal anti-inflammatory drugs on thedisposition of methotrexate in patients with rheumatoid arthritis. Biopharm. Drug

    Dispos. 19: 163167.

    46. Mawatari, H., K. Unei, S. Nishimura, N. Sakura, and K. Ueda. 2001. Compar-ative pharmacokinetics of oral 6-mercaptopurine and intravenous 6-mercaptopu-rine riboside in children. Pediatr. Int. 43: 673677.

    47. Lieberman, M. M., G. M. Patterson, and R. E. Moore. 2001. In vitro bioassays foranticancer drug screening: effects of cell concentration and other assay parame-ters on growth inhibitory activity. Cancer Lett. 173: 2129.

    48. Tichopad, A., M. Dilger, G. Schwarz, and M. W. Pfaffl. 2003. Standardizeddetermination of real-time PCR efficiency from a single reaction set-up. Nucleic

    Acids Res. 31: e122.

    49. Green, K. J. 2002. Improving understanding of exercise effects on in vitro T-lymphocyte functionthe role of fluorescent cell division tracking. Exerc. Im-munol. Rev. 8: 101115.

    50. Mannering, S. I., J. Morris, K. Jensen, A. Purcell, M. Honeyman, P. van Endert,and L. Harrison. 2003. A sensitive method for detecting proliferation of rareautoantigen-specific human T cells. J. Immunol. Methods 283: 173183.

    51. Thompson, B. S., and T. C. Mitchell. 2004. Measurement of daughter cell accu-mulation during lymphocyte proliferation in vivo. J. Immunol. Methods 295:7987.

    5888 ABNORMAL PHENOTYPE AND FUNCTION OF LUPUS DCs

    p

    ,

    j

    g

    http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/
  • 7/30/2019 Sle Review

    13/13

    52. Chen, J. C., M. L. Chang, and M. O. Muench. 2003. A kinetic study of the murinemixed lymphocyte reaction by 5,6-carboxyfluorescein diacetate succinimidyl es-ter labeling. J. Immunol. Methods 279: 123133.

    53. Renno, T., A. Attinger, S. Locatelli, T. Bakker, S. Vacheron, andH. R. MacDonald. 1999. Cutting edge: apoptosis of superantigen-activated Tcells occurs preferentially after a discrete number of cell divisions in vivo. J. Im-munol. 162: 63126315.

    54. Koller, M., B. Zwolfer, G. Steiner, J. Smolen, and C. Scheinecker. 2004. Phe-notypic and functional deficiencies of monocyte-derived dendritic cells in sys-temic lupus erythematosus (SLE) patients. Int. Immunol. 16: 15951604.

    55. Cao, W., S. Lee, and J. Lu. 2004. CD83 is preformed inside monocytes, macro-phages and dendritic cells but it is only stably expressed on activated dendritic

    cells. Biochem. J. 385: 8593.56. Adachi, Y., S. Taketani, J. Toki, K. Ikebukuro, K. Sugiura, H. Oyaizu,

    R. Yasumizu, M. Tomita, H. Kaneda, Y. Amoh, et al. 2002. Marked increase innumber of dendritic cells in autoimmune-prone (NZW BXSB)F

    1mice with

    age. Stem Cells 20: 6172.57. Scheinecker, C., B. Zwolfer, M. Koller, G. Manner, and J. Smolen. 2001. Alter-

    ations of dendritic cells in systemic lupus erythematosus: phenotypic and func-tional deficiencies. Arthritis Rheum. 44: 856 865.

    58. Nakajima, A., M. Azuma, S. Kodera, S. Nuriya, A. Terashi, M. Abe, S. Hirose,T. Shirai, H. Yagita, and K. Okumura. 1995. Preferential dependence of autoan-tibody production in murine lupus on CD86 costimulatory molecule. Eur. J. Im-munol. 25: 30603069.

    59. Kinoshita, K., G. Tesch, A. Schwarting, R. Maron, A. Sharpe, and V. Kelley.2000. Costimulation by B7-1 and B7-2 is required for autoimmune disease inMRL-Faslpr mice. J. Immunol. 164: 60466056.

    60. Georgiev, M., L. Agle, J. Chu, K. Elkon, and D. Ashany. 2005. Mature dendriticcells readily break tolerance in normal mice but do not lead to disease expression.

    Arthritis Rheum. 52: 225238.

    61. Holcombe, R. F., B. Baethge, R. Wolf, K. Betzing, R. Stewart, V. Hall, andM. Fukuda. 1994. Correlation of serum interleukin-8 and cell surface lysosome-associated membrane protein expression with clinical disease activity in systemiclupus erythematosus. Lupus 3: 97102.

    62. Rovin, B. H., L. Lu, and X. Zhang. 2002. A novel interleukin-8 polymorphism isassociated with severe systemic lupus erythematosus nephritis. Kidney Int. 62:261265.

    63. Tsai, C. Y., T. H. Wu, C. Yu, J. Lu, and Y. Tsai. 2000. Increased excretions of2-microglobulin, IL-6, and IL-8 and decreased excretion of Tamm-Horsfall gly-coprotein in urine of patients with active lupus nephritis. Nephron 85: 207214.

    64. Scimone, M. L., V. P. Lutzky, S. Zittermann, P. Maffia, C. Jancic, F. Buzzola,A. Issekutz, and H. Chuluyan. 2005. Migration of polymorphonuclear leucocytesis influenced by dendritic cells. Immunology 114: 375385.

    65. Trysberg, E., H. Carlsten, and A. Tarkowski. 2000. Intrathecal cytokines in sys-temic lupus erythematosus with central nervous system involvement. Lupus 9:498503.

    66. Zhu, K., Q. Shen, M. Ulrich, and M. Zheng. 2000. Human monocyte-deriveddendritic cells expressing both chemotactic cytokines IL-8, MCP-1, RANTES

    and their receptors, and their selective migration to these chemokines. Chin. Med.J. 113: 11241128.

    67. Mayer, G., K. Pohlmeyer, A. Caliebe, E. Heimueller, B. Behnke, G. Steimann,C. Lange, and J. Beuth. 2000. Low molecular thymic peptides stimulate humanblood dendritic cells. Anticancer Res. 20: 28732883.

    68. Bickel, B. M. 1993. The role of interleukin-8 in inflammation and mechanisms ofregulation. J. Periodontol. 64(Suppl. 5): 456460.

    69. Huang, S., L. Mills, B. Mian, C. Tellez, M. McCarty, X. Yang, J. Gudas, andM. Bar-Eli. 2002. Fully humanized neutralizing antibodies to interleukin-8(ABX-IL8) inhibit angiogenesis, tumor growth, and metastasis of human mela-noma. Am. J. Pathol. 161: 125134.

    70. Mukaida, N. 2000. Interleukin-8: an expanding universe beyond neutrophil che-motaxis and activation. Am. J. Hematol. 72: 391398.

    71. Gerszten, T. E., E. Garcia-Zepeda, Y. Lim, M. Yushida, H. Ding, M. Gimbrone,A. Luster, F. Luscinskas, and A. Rosenzweig. 1999. MCP-1 and IL-8 trigger firmadhesion of monocytes to vascular endothelium under flow conditions. Nature398: 718723.

    72. Casilli, F., A. Bianchini, I. Gloaguen, L. Biordi, E. Alesse, C. Festuccia,B. Cavalieri, R. Strippoli, M. Cervellera, R. Di Bitondo, et al. 2005. Inhibition ofinterleukin-8 (CXCL8/IL-8) responses by repertaxin, a new inhibitor of the che-mokine receptors CXCR1 and CXCR2. Biochem. Pharmacol. 69: 385394.

    73. Lai, K., J. Leung, K. Lai, and C. Lai. 1997. Effect of anti-DNA autoantibodies onthe gene expression of interleukin 8, transforming growth factor-, and nitricoxide synthase in cultured endothelial cells. Scand. J. Rheumatol. 26: 461467.

    74. Sun, K. H., C. Yu, S. Tang, and G. Sun. 2000. Monoclonal anti-double-strandedDNA autoantibody stimulates the expression and release of Il-1, Il-6, IL-8 IL-10and TNF- from normal human mononuclear cells involving in the lupus patho-genesis. Immunology 99: 352360.

    75. Rus, V., S. Atamas, V. Shustova, I. Luzina, F. Selaru, L. Madger, and C. Via.2002. Expression of cytokine- and chemokine-related genes in peripheral bloodmononuclear cels from lupus patients by cDNA array. Clin. Immunol. 102:283290.

    76. Scholler, N., M. Hayden-Ledbetter, K. Hellstrom, I. Hellstrom, and J. Ledbetter.2001. CD83 is a sialic acid-bindingIg-like lectin(Siglec) adhesion receptor that bindsmonocytes and a subset of activated CD8 T cells. J. Immunol. 166: 38653872

    77. Kruse, M., O. Rosorius, F. Kratzer, D. Bevec, C. Kuhnt, A. Steinkasserer,G. Schuler, and J. Hauber. 2000. Inhibition of CD83 cell surface expression

    during dendritic cell maturation by interference with nuclear export of CD83mRNA. J. Exp. Med. 191: 15811590.

    78. Lechmann, M., E. Zinser, A. Golka, and A. Steinkasserer. 2002. Role of CD83in the immunomodulation of dendritic cells. Int. Arch. Allergy Immunol. 129:113118.

    79. Lechmann, M., E. Kremmer, H. Sticht, and A. Steinkasserer. 2002. Overexpression,purification, and biochemical characterization of the extracellular human CD83 do-main and generation of monoclonal antibodies. Protein Expr. Purif. 24: 445452.

    80. Scholler, N., M. Hayden-Ledbetter, A. Dahlin, I. Hellstrom, K. Hellstrom, andJ. Ledbetter. 2002. Cutting edge: CD83 regulates the development of cellularimmunity. J. Immunol. 168: 25992602.

    81. Hirano, N., M. O. Butler, Z. Xia, S. Ansen, M. S. von Bergwelt-Baildon,D. Neuberg, G. J. Freeman, and L. M. Nadler. 2005. Engagement of CD83ligand induces prolonged expansion of CD8 T cells and preferential enrich-ment for antigen specificity. Blood 107: 15281536.

    82. Filaci, G., S. Bacilieri, M. Fravega, M. Monetti, P. Contini, M. Ghio, M. Setti,F. Puppo, and F. Indiveri. 2001. Impairment of CD8 T suppressor cell function inpatients with active systemic lupus erythematosus. J. Immunol. 166: 64526457.

    83. Garcia-Martinez, L., M. Appleby, K. Staehling-Hampton, D. Andrews, Y. Chen,M. McEuen, P. Tang, R. Rhinehart, S. Proll, B. Paeper, et al. 2004. A novelmutation in CD83 results in the development of a unique population of CD4

    T cells. J. Immunol. 173: 29953001.

    84. Schwab, M., and U. Klotz. 2001. Pharmacokinetic considerations in the treat-ment of inflammatory bowel disease. Clin. Pharmacokinet. 40: 723751.

    85. Zhu, J., X. Liu, C. Xie, M. Yan, Y. Yu, E. S. Sobel, E. K. Wakeland, andC. Mohan. 2005. T cell hyperactivity in lupus as a consequence of hyperstimu-latory antigen-presenting cells. J. Clin. Invest. 115: 18691878.

    86. Oren, A., C. Husebo, A. C. Iversen, and R. Austgulen. 2005. A comparativestudy of immunomagnetic methods used for separation of human natural killercells from peripheral blood. J. Immunol. Methods 303: 110.

    87. Tobiasova-Czetoova, Z., A. Palmborg, A. Lundqvist, G. Karlsson, L. Adamson,J. Bartunkova, G. Masucci, and P. Pisa. 2005. Effects of human plasma proteins onmaturation of monocyte-derived dendritic cells. Immunol. Lett. 100: 113119.

    88. Tkachenko, N., K. Wojas, J. Tabarkiewicz, and J. Rolinski. 2005. Generation ofdendritic cells from human peripheral blood monocytescomparison of differ-ent culture medium. Folia Histochem. Cytobiol. 43: 2530.

    89. Kufner, S., H. Zitzelsberger, T. Kroell, R. Pelka-Fleischer, A. Salem, F. de Valle,C. Schweiger, V. Nuessler, C. Schmid, H. J. Kolb, and H. M. Schmetzer. 2005.Leukemia-derived dendritic cells can be generated from blood or bone marrow cellsfrom patients with acute myeloid leukaemia: a methodological approach under se-rum-free culture conditions. Scand. J. Immunol. 62: 8698.

    90. Chernysheva, A. D., K. A. Kirou, and M. K. Crow. 2002. T cell proliferationinduced by autologous non-T cells is a response to apoptotic cells processed bydendritic cells. J. Immunol. 169: 12411250.

    91. Banki, Z., L. Kacani, B. Mullauer, D. Wilflingseder, G. Obermoser,H. Niederegger, H. Schennach, G. Sprinzl, N. Sepp, A. Erdei, et al. 2003. Cross-linking of CD32 induces maturation of human monocyte-derived dendritic cellsvia NF-B signaling pathway. J. Immunol. 170: 39633970.

    92. Radstake, T. R., K. C. Nabbe, M. H. Wenink, M. F. Roelofs, A. Oosterlaar,A. W. van Lieshout, P. Barrera, P. L. van Lent, and W. B. van den Berg. 2005.Dendritic cells from patients with rheumatoid arthritis lack the interleukin 13mediated increase of FcRII expression, which has clear functional conse-quences. Ann. Rheum. Dis. 64: 17371743.

    93. Radstake, T. R., A. B. Blom, A. W. Sloetjes, E. O. van Gorselen, G. J. Pesman,L. Engelen, R. Torensma, W. B. van den Berg, C. G. Figdor, P. L. van Lent, etal. 2004. Increased FcRII expression and aberrant tumour necrosis factor production by mature dendritic cells from patients with active rheumatoid ar-thritis. Ann. Rheum. Dis. 63: 15561563.

    94. Roelofs, M. F., L. A. Joosten, S. Abdollahi-Roodsaz, A. W. van Lieshout,T. Sprong, F. H. van den Hoogen, W. B. van den Berg, and T. R. Radstake.2005. The expression of Toll-like receptors 3 and 7 in rheumatoid arthritissynovium is increased and costimulation of Toll-like receptors 3, 4, and 7/8results in synergistic cytokine production by dendritic cells. Arthritis Rheum. 52:23132322.

    95. Tran, C. N., S. K. Lundy, and D. A. Fox. 2005. Synovial biology and T cells inrheumatoid arthritis. Pathophysiology 12: 183189.

    96. Klareskog, L., U. Forsum, A. Scheynius, D. Kabelitz, and H. Wigzell. 1982.Evidence in support of a self-perpetuating HLA-DR-dependent delayed-type

    cell reaction in rheumatoid arthritis. Proc. Natl. Acad. Sci. USA 79: 36323636.97. Palucka, A. K., J. P. Blanck, L. Bennett, V. Pascual, and J. Banchereau. 2005.

    Cross-regulation of TNF and IFN- in autoimmune diseases. Proc. Natl. Acad.Sci. USA 102: 33723377.

    98. Kyttaris, V. C., Y. T. Juang, and G. C. Tsokos. 2005. Immune cells and cyto-kines in systemic lupus erythematosus: an update. Curr. Opin. Rheumatol. 17:518522.

    99. Lorenz, H. M., M. Herrmann, and J. R. Kalden. 2001. The pathogenesis ofautoimmune diseases. Scand. J. Clin. Lab. Invest. Suppl. 235: 1626.

    100. Vallin, H., A. Perers, G.V. Alm, and L. Ronnblom. 1999. Anti-double-strandedDNA antibodies and immunostimulatory plasmid DNA in combination mimicthe endogenous IFN- inducer in systemic lupus erythematosus. J. Immunol.163: 63066313.

    101. Boule, M., C. Broughton, F. Mackay, S. Akira, A. Marshak-Rothstein, andI. Rifkin.2004. Toll-like receptor 9-dependent and -independent dendritic cell activation bychromatin-immunoglobulin G complexes. J. Exp. Med. 199: 16311640.

    102. Decker, P., H. Singh-Jasuja, S. Haager, I. Kotter, and H. Rammensee. 2005.Nucleosome, the main autoantigen in systemic lupus erythematosus, induces

    direct dendritic cell activation via a MyD88-independent pathway: conse-quences on inflammation. J. Immunol. 174: 33263334.

    5889The Journal of Immunology

    p

    ,

    j

    g

    http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/http://www.jimmunol.org/