chelvarajan - leukocyte biol 06 - aging micro array

Upload: f1r3br4nd9427

Post on 07-Apr-2018

218 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/6/2019 Chelvarajan - Leukocyte Biol 06 - Aging Micro Array

    1/14

    Molecular basis of age-associated cytokine dysregulation in

    LPS-stimulated macrophages

    R. Lakshman Chelvarajan,*, Yushu Liu, Diana Popa,* Marilyn L. Getchell,*,

    Thomas V. Getchell,*,, Arnold J. Stromberg, and Subbarao Bondada*,,1

    *Sanders Brown Center on Aging and Departments of Microbiology, Immunology & Molecular Genetics, Statistics,Anatomy and Neurobiology, and Physiology, University of Kentucky, Lexington

    Abstract: Aged humans and rodents are suscepti-ble to infection with Streptococcus pneumoniaebacteria as a result of an inability to make antibod-ies to capsular polysaccharides. This is partly aresult of decreased production of proinflammatorycytokines and increased production of interleukin(IL)-10 by macrophages (M) from aged mice. Tounderstand the molecular basis of cytokine dys-regulation in aged mouse M, a microarray anal-ysis was performed on RNA from resting and lipo-polysaccharide (LPS)-stimulated M from agedand control mice using the Affymetrix Mouse Ge-nome 430 2.0 gene chip. Two-way ANOVA analy-sis demonstrated that at an overall P < 0.01 level,853 genes were regulated by LPS (169 in only theyoung, 184 in only the aged, and 500 in both).Expression analysis of systematic explorer re-vealed that immune response (proinflammatorychemokines, cytokines, and their receptors) andsignal transduction genes were specifically reducedin aged mouse M. Accordingly, expression of Il1and Il6 was reduced, and Il10 was increased, con-

    firming our previous results. There was also de-creased expression of interferon-. Genes in theToll-like receptor-signaling pathway leading to nu-clear factor-B activation were also down-regu-lated but IL-1 receptor-associated kinase 3, a neg-ative regulator of this pathway, was increased inaged mice. An increase in expression of the genefor p38 mitogen-activated protein kinase (MAPK)was observed with a corresponding increase in pro-tein expression and enzyme activity confirmed byWestern blotting. Low doses of a p38 MAPK inhib-itor (SB203580) enhanced proinflammatory cyto-

    kine production by M and reduced IL-10 levels,indicating that increased p38 MAPK activity has arole in cytokine dysregulation in the aged mouseM. J. Leukoc. Biol. 79: 13141327; 2006.

    Key Words: inflammation microarray p38 MAPK

    INTRODUCTION

    Function of the immune system is decreased with age, leading

    to increased susceptibility of the elderly to infections [1, 2],

    and infections with influenza viruses and Streptococcus pneu-

    moniae, which lead to pneumonia, are a major cause of mor-

    bidity and mortality in the elderly. Previous studies have

    suggested that influenza virus-specific T cell responses are

    decreased in the aged, and that it is in part a result of defects

    in antigen presentation [3]. The increased incidence of pneu-

    mococcal infections is a result of a defect in the production of

    antibodies to the capsular polysaccharide antigens, which are

    critical for killing of the bacteria by the phagocytic cells [4].

    The antibody response to polysaccharides is dependent on Bcells and macrophages (M), and our recent studies suggest

    the defects in the aged are in part a result of deficiencies in

    M function [5].

    Pure polysaccharide vaccines are less effective in aged

    individuals compared with young adults [4, 6]. Neonates are

    also unresponsive to polysaccharide antigens or vaccines based

    on pure polysaccharides. Although the cellular basis of this

    unresponsiveness in neonates has been attributed to B cell

    immaturity, we have found that neonatal M are defective in

    supporting B cell responses from young mice to polysaccharide

    antigens [79]. In the aged, where mature B cells are present

    and are only marginally affected in their responses to B cell

    receptor signaling, the reason for reduced responsiveness to

    polysaccharide vaccines is not known. The pneumococcal

    polysaccharides have been classified as thymic-independent

    antigens, as they do not elicit antigen-specific helper T cells,

    which can interact with B cells in a cognate manner [10].

    However, B cells and M are required for generating an

    immune response to polysaccharide antigens. We have focused

    on the spleen, as it is known that spleen is critical for immune

    responses against polysaccharide-encapsulated bacteria [11].

    B cells and marginal zone M from the spleen have a role in

    the antibody response to polysaccharide antigens [12, 13].

    Recently, using a mouse model system, we have shown that

    splenic M from young but not the aged mice are able tosupport young mouse B cells to produce antipolysaccharide

    antibodies [1]. In this system, we showed that the main function

    of M is to produce the cytokines interleukin (IL)-1 and IL-6,

    which are needed for B cell differentiation. In fact, in the

    presence of added IL-1 and IL-6 B cells from the young and the

    1 Correspondence: Sanders Brown Center on Aging, Room 329, University of

    Kentucky, Lexington, KY 40536. E-mail: [email protected]

    Received January 12, 2006; revised February 11, 2006; accepted February

    20, 2006; doi: 10.1189/jlb.0106024.

    1314 Journal of Leukocyte Biology Volume 79, June 2006 0741-5400/06/0079-1314 Society for Leukocyte Biology

  • 8/6/2019 Chelvarajan - Leukocyte Biol 06 - Aging Micro Array

    2/14

    aged produced equivalent levels of antipolysaccharide re-

    sponses. A major reason for the inability of M from the aged

    to support B cell responses to polysaccharide antigens was a

    result of a defect in secretion of IL-1 and IL-6. However, the

    cytokine secretion defect was not limited to IL-1 and IL-6, as

    other proinflammatory cytokines, such as IL-12 and tumor

    necrosis factor (TNF-) were also produced at lower levels

    by M from the aged in comparison with young mice. It is

    interesting that the M from the aged were not defective in

    IL-10 production but produced more of this cytokine than M

    from the young. Thus, the cytokine production was dysregu-

    lated in M from the aged.

    Similar defects in production of IL-1, IL-6, TNF-, M-

    inflammatory protein (MIP)-1 and MIP-1 by M from aged

    mice have been noted by several other researchers [1417].

    Defects in M function, in particular, a reduction in secretion

    of vascular endothelial growth factor and expression of cell

    adhesion molecules, are thought to contribute to the delay in

    wound healing in the aged [18]. In contrast, peritoneal M

    from aged mice have been shown to produce more cyclooxy-

    genase-2 (COX-2) and prostaglandin E2 (PGE2) in response to

    lipopolysaccharide (LPS) stimulation [19]. Renshaw et al. [15]

    found that expression of a variety of Toll-like receptors (TLRs),

    including TLR4, was decreased in the aged, which could be the

    reason for a decreased response of M from aged mice to LPS.

    However, this does not explain increased IL-10 production in

    the ageda cytokine not examined by Renshaw et al. [15].

    Conversely, Boehmer et al. [20, 21] found a decrease in pro-

    duction of TNF- and IL-6 by thioglycollate-elicited peritoneal

    M or splenic M from the aged but did not find a reduction

    in TLR4 expression. Instead, they attributed the reduced cy-

    tokine production to a decrease in c-jun N-terminal kinase

    (JNK) and p38 mitogen-activated protein kinase (MAPK) ac-

    tivation in M from the aged. Few studies have measured the

    effect of age on M in a comprehensive manner.To determine if cytokine dysregulation in the splenic M

    from aged mice extends to other proinflammatory cytokines and

    chemokines and to understand the molecular basis of the

    altered function of M from aged mice, we performed a mi-

    croarray analysis of genes expressed in M from young and

    aged mice stimulated with LPS. Despite many microarray

    studies on M [2226], few studies focused on splenic M,

    and none have compared the transcriptional profile of M as a

    function of age. Our study is thus unique in that it provides

    novel information about gene expression in splenic M from

    young and the aged mice. This allowed us to test several

    hypotheses to interpret the age-associated dysfunction in M.This analysis confirmed and extended to the gene level our

    findings about cytokine dysregulation and showed that several

    other proinflammatory cytokines and chemokines were de-

    creased in the aged, suggesting that decreased TLR signaling

    may be involved in the M defect. It is most interesting that

    the microarray data showed that the expression of the gene

    encoding for p38 MAPK was increased dramatically in the

    M from aged mice, which was confirmed by Western blot

    analysis. Moreover, the increased p38 MAPK activity appeared

    to have a causal role in the cytokine dysregulation phenotype

    of M from aged mice.

    MATERIALS AND METHODS

    Mice

    Female young (4 months old) and aged (2022 months old) BALB/c mice were

    obtained from the colonies of the National Institute on Aging, National Insti-

    tutes of Health (NIH; Bethesda, MD). The mice were maintained in the animal

    facility at the Department of Laboratory Animal Research on a 12:12 h

    light/dark cycle and were given food and water ad libitum. All protocols were

    implemented in accordance with NIH guidelines and approved by the Univer-

    sity of Kentucky Institutional Animal Care and Use Committee (Lexington).

    Reagents

    SB203580, the inhibitor of p38 MAPK, was obtained from Calbiochem (San

    Diego, CA). Antibodies against the MAPKs were obtained from Cell Signaling

    Technologies (Beverly, MA), and the monoclonal antibody against -actin was

    obtained from Sigma Chemical Co. (St. Louis, MO). Gel-purified LPS (Esche-

    richia coli 055:B5) was obtained from Sigma Chemical Co., and fluorochrome-

    conjugated antibody to CD11b (membrane-activated complex-1 or Mac-1) was

    purchased from Caltag (Burlingame, CA).

    Cell preparation

    CD11b-positive cells were purified from spleens of mice using CD11b anti-

    body-coupled magnetic beads (Miltenyi Biotec, Bergisch Gladbachuor, Ger-

    many). The purified cells were found to be routinely 9095% CD11b-positive

    when tested by flow cytometry. For each experiment, the CD11b-positive cellswere pooled from two young and two aged mice. However, for the microarray

    study, the CD11b-positive cells were pooled from 10 young and five aged mice.

    Cell culture

    For the microarray study, M (10106) were cultured in triplicate in Iscove/

    F12 medium 10% fetal calf serum at 37C in 5% CO2 at a density of 1

    106/ml. Cultures were stimulated with 1 g/ml LPS or left unstimulated for 6 h.

    RNA isolation

    The cells were spun down in the plate, the growth medium removed by

    pipetting, and the cultures were incubated for 5 min in TRI Reagent (Molec-

    ular Research Center, Inc., Cincinnati, OH). Total RNA was extracted under

    RNase-free conditions and was further purified using the Qiagen RNeasy mini

    kit, according to the manufacturers protocol (Qiagen, Valencia, CA). TotalRNA yield and purity were assessed with a spectrophotometer and with the

    Model 2100 bioanalyzer (Agilent Technologies, Palo Alto, CA); all samples

    had two sharp peaks corresponding to 18S and 28S RNA on the bioanalyzer

    electropherograms. The RNA was then transcribed into cRNA by the microar-

    ray facility and was hybridized to the whole mouse genome chips (Affymetrix

    Mouse Genome 430 2.0) using one chip for RNA from each culture, resulting

    in a total of 12 gene chips.

    Microarray data analysis

    From each sample, 4 g total RNA was used for the amplification and labeling

    reactions. Out of this, 20 g labeled cRNA was used in the hybridization

    reaction. The Affymetrix Mouse Genome 430 2.0 has 45,101 probe sets, and

    the probe sets with absent calls across all samples and unannotated genes were

    removed to reduce the multiple testing problem. These steps resulted in 18,627

    probe sets. Two-way ANOVA tests were carried out to identify differentially

    expressed genes. For each probe set, the model yijk i j ij

    ijk was applied, where yijk is the log-transformed expression level of the kth

    chip in the ith age and the jth LPS. Furthermore, represents the grand mean

    expression, i is the effect as a result of age [aged (A) and young (Y)], j is the

    effect as a result of the LPS [medium (M) and LPS (L)], ij is the interaction

    effect between age and LPS, and ijk is an error term, which is assumed to be

    normally distributed with mean 0 and variance 2. Applying an overall P value

    of 0.01, ANOVA analysis indicated that only 9894 out of the 18,627 probe

    sets were significantly regulated for age, treatment, or both. To find the genes

    with biological significance, we applied the intensity, fold-change, and pair-

    wise P values as additional filters. Probe sets with mean intensities 200

    across all four treatments or fold changes that were less than twofold and

    pair-wise P value (intensity with LPS vs. that for medium) 0.01 were not

    Chelvarajan et al. Macrophages from aged mice are defective and have increased p38 MAPK 1315

  • 8/6/2019 Chelvarajan - Leukocyte Biol 06 - Aging Micro Array

    3/14

    analyzed further. Application of these filters resulted in 1115 probe sets. As

    the gene chip has a large number of genes detected by multiple probe sets, to

    produce the Venn diagram (see Fig. 2), principal component analysis was

    applied to resolve genes with multiple probe sets that appear in more than one

    region of the Venn diagram. After principal component analysis, the number of

    unique genes regulated by LPS was further reduced to 853.

    The gene-tree diagram was constructed using the GeneSpring 7.2 (Agilent

    Technologies) program. Expression analysis of systematic explorer (EASE) was

    used to facilitate the biological interpretation of gene lists derived from the

    results of the microarray and was accessed online from the Database for

    Annotation, Visualization and Integrated Discovery website (http://

    apps1.niaid.nih.gov/david/) from the National Institute of Allergy and Infec-

    tious Disease, NIH.

    Real-time reverse transcriptase-polymerasechain reaction (RT-PCR)

    For RT-PCR, the 12 RNA samples used for Microarray plus a further six

    samples were reverse-transcribed using the High Capacity cDNA Archive kit

    (Applied Biosystems, Foster City, CA). The cDNA was then amplified by

    real-time PCR using TaqMan primers, probes, and TaqMan Master Mix on an

    ABI Prism 7000 sequence detection system (Applied Biosystems). The Taq-

    Man primers and probes used were designed by Applied Biosystems to cover

    the intron-exon junction of the respective genes and were tested further by the

    manufacturer to prove their specificity for the genes for which they were

    designed. The standard curve was done in duplicate from cDNA derived from

    P388D1 cells stimulated with LPS. The experimental cDNA was run in

    triplicate and normalized to 18S RNA, and the no-template controls were donein duplicate.

    Cytokine analysis

    M (0.25106) were cultured in duplicate for 1 day in 1 g/ml LPS, various

    doses of SB203580, or medium only. Various cytokines in the supernatant were

    estimated in duplicate using enzyme-linked immunosorbent assay (ELISA).

    IL-12, IL-10, and TNF- were estimated with OptEIA kits (PharMingen, San

    Diego, CA). IL-6 was measured with a matched-pair antibody set (Clones

    MP5-20F3 and MP5-32C11) from BD Biosciences (San Jose, CA). The optical

    densities (OD) were read on an HTS 7000 (Perkin Elmer, Norwalk, CT).

    Results are presented as mean SE of four measurements.

    Western blotting

    Approximately 1.5 106 M were cultured per well per 24-well plate. After

    allowing the cells to rest for at least 90 min, the cells were stimulated with LPS

    for 15 min. Cells were lysed in the plate using the lysis buffer provided by Cell

    Signaling Technologies. An aliquot of the lysate was subjected to sodium

    dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and Western

    blot analysis. The blots were analyzed by probing the membrane using various

    primary antibodies followed by horseradish peroxidase-conjugated secondary

    antibodies (Santa Cruz Biotechnologies, CA). The blots were developed with

    the Pico Chemiluminescence substrate (Pierce Biotechnology, Rockford, IL)

    and exposed to Kodak X-Omat film, which was then scanned by a Kodak Image

    Station 2000RT (Eastman Kodak, New Haven, CT). For reprobing, membranes

    were stripped using a solution containing 62.5 mM tris(hydroxymethyl)amino-

    methane-HCl, 2% SDS, and 100 mM -mercaptoethanol at 65C for 5 min. The

    relative integrated OD of the protein bands was estimated using the Kodak

    Image Station. Band intensities were normalized by dividing the intensity of

    phosphorylated protein by that of total protein [e.g., for extracellular signal-regulated kinase (ERK)1/2] or by dividing protein of interest by that of-actin

    (e.g., for p38 MAPK).

    Statistical analysis

    Students t test was used to evaluate the significance of the differences among

    means in Western blots, ELISA data, and PCR data.

    RESULTS

    Definition of LPS and aging signatures

    The Affymetrix Mouse Genome 430 2.0 whole mouse genome

    gene chip, which was used in this study, has 45,101 probe sets.

    After removing the probe sets for unannotated and control

    genes and expression sequence tags, a two-way ANOVA anal-

    ysis determined that 9894 probe sets were regulated differen-

    tially between the two age groups (overall P value 0.01). To

    get a broad overview of the gene expression pattern changes

    with age and/or LPS stimulation, a gene tree was created using

    GeneSpring, which then revealed a number of patterns of gene

    expression (Fig. 1). Region A indicates a set of genes that were

    induced upon stimulation with LPS in M from the aged and

    young. These genes had low constitutive levels of expression

    and upon stimulation, were of comparable intensities in M

    from young and aged. Genes, which were present at low inten-

    sities in unstimulated M but were induced by LPS in only

    Fig. 1. A gene tree of the probe sets from the overall P 0.01 list. A

    microarray analysis was performed on M from young and aged mice cultured

    for 6 h with LPS. For each treatment, RNA was obtained from triplicate M

    cultures (each culture was processed separately) and hybridized onto separate

    gene chips. Depicted here are the 9894 probe sets, which were identified to be

    differentially expressed when the overall P value was 0.01 (two-way ANOVA

    analysis). Each column represents the expression levels for a given gene chip.

    Using the GeneSpring computer program, we applied a hierarchical method to

    cluster these probe sets. Pearson correlation coefficient was used as the

    distance measure. The analysis shows clusters of genes regulated by age only,

    LPS only, or by both, identified as Regions AF, and explained in the text. The

    color-coding is for the intensity of expression of each probe set, and red is the

    highest; green, the lowest.

    1316 Journal of Leukocyte Biology Volume 79, June 2006 http://www.jleukbio.org

  • 8/6/2019 Chelvarajan - Leukocyte Biol 06 - Aging Micro Array

    4/14

    M from young mice or from aged mice, are depicted in

    Regions B and E, respectively. The expression of genes de-

    picted in Regions C and D were not modulated by LPS. Those

    in Region C were significantly higher in M from young mice,

    and those in Region D were higher in M from the aged.

    Collectively, these genes would constitute an aging signature

    and include pellino 1 (Peli1; Region C) and chemokine-binding

    protein 2 (Ccbp2), Mapk14, and IL-1 receptor (IL-1R)-associ-

    ated kinase (Irak)3 and several genes related to proliferation

    (Region D). Genes in Region F were expressed constitutively at

    high levels in both age groups but are dramatically down-

    regulated upon LPS stimulation [e.g., peroxisome proliferator-

    activated receptor- (Ppar) and CC chemokine ligand 24

    (Ccl24)].

    Analysis of the effect of age on genes affectedby LPS stimulation

    To begin the task of identifying the key genes involved in the

    differential response between M from aged and young, probe

    sets with low mean intensities200 in all four treatments (i.e.,

    youngLPS and agedLPS) were eliminated, and then a cri-

    terion of at least a twofold change was used for all pair-wise

    comparisons (i.e., LPS and/or age). The remaining probe setswere then divided further into those that were uniquely regu-

    lated in M from the aged, young, or both. As a number of

    genes are detected by multiple probe sets on the Affymetrix

    Mouse Genome 430 2.0 gene chip, the probe set numbers were

    converted to the numbers of genes. Where genes were detected

    by probe sets with conflicting patterns of regulation, principal

    component analysis was used to assign these genes to one

    pattern. The numbers of genes regulated by LPS were then

    summarized in a Venn diagram (Fig. 2). A total of 853 genes

    was affected by LPS stimulation. A comparable number of

    genes were uniquely regulated in M from the young and aged

    (169 and 184 genes, respectively), and the remainder (500) wasregulated in both ages by LPS. It is interesting that more genes

    were down-regulated than up-regulated by LPS in either age

    group: In all, 500 genes (or 60%) were down-regulated in one

    age group or the other or both age groups, and expression of the

    remaining 40% was up-regulated by LPS (Fig. 2). The common

    subset (i.e., genes affected by LPS in both age groups) was

    divided further into four groups, of which 194 were increased

    in M from young and aged, and 297 were decreased in both

    groups of M. Another interesting feature of the response to

    LPS is that in the common subset, there were few genes (nine)

    that were cross-regulated (i.e., up in the age and down in the

    young and vice versa).

    Validation of microarray data by ELISA

    We had previously shown by ELISA that aged M secreted

    lower amounts of the proinflammatory cytokines IL-1, IL-6,

    IL-12, and TNF- but higher amounts of IL-10 than M from

    the young [1]. These cytokines were also affected similarly in

    the microarray analysis (i.e., reduced amounts of mRNA for

    IL-1, IL-6, IL-12, and TNF- and increased levels of IL-10

    mRNA). Thus, the altered pattern of cytokine secretion ob-

    served with M from the aged is also reflected at the steady-

    state levels of mRNA for these cytokines and therefore, vali-

    dating the microarray dataset for these key pro- and anti-

    inflammatory cytokines (Fig. 3 and data not shown). It is to be

    noted that ELISA data show that IL-6 is the most abundant

    cytokine, and IL-1 is produced at the lowest level in M from

    either age group. However, this is not reflected by the intensity

    data from the microarray (IL-1 gives the highest signal), which

    is because the hybridization efficiencies are different across

    the various probe sets on the chips.

    EASE: cytokine and chemokine subset

    To identify functional categories of genes that could be respon-

    sible for the difference in cytokine secretion, we divided theprobe sets from the overall P 0.01 list into those that were

    at least twofold higher or lower (aged vs. young) upon LPS

    stimulation. We then performed an EASE of the LPS-regulated

    genes identified by the microarray data [27]. EASE calculates

    EASE scores to identify over-represented gene categories

    within lists of genes. Using a cut-off value of an EASE score

    0.001 for significance, we have found that a variety of im-

    mune-response genes (including cytokines and chemokines)

    and signal transduction genes (TLR and MAPK pathways) was

    over-represented among the probe sets that were lower in M

    from aged when compared with M from the young (data not

    shown). We then analyzed the expression of the genes encoding

    cytokines and chemokines from our microarray data (Table 1).Although a majority of the cytokines and chemokines depicted

    is lower in M from the aged, a few, such as Ccbp2, are

    expressed at considerably higher amounts in M from the

    aged, before and after LPS stimulation. A number of genes

    [e.g., CXC chemokine ligand 16 (Cxcl16)] were higher in M

    from the young, and this difference remains even upon LPS

    stimulation. A number of genes, such as Ccl24, were actually

    turned off by LPS.

    The microarray data show that in addition to the cytokines

    identified by us by ELISA assays, IFN-, CSF-1, GM-CSF, and

    bone morphogenetic protein-1 (BMP-1; Ifng, Csf1, Csf2, and

    Fig. 2. A Venn diagram of the genes regulated by LPS in M from young and

    aged mice. Only probe sets with a pair-wise P 0.01 and an LPS-induced fold

    change of at least two (up- and down-regulation) were considered. Where there

    were multiple probe sets for the same gene in more than one region of the Venn

    diagram, principal component analysis, a dimension-reduction technique, was

    used to reduce the number of probe sets to unique genes.

    Chelvarajan et al. Macrophages from aged mice are defective and have increased p38 MAPK 1317

  • 8/6/2019 Chelvarajan - Leukocyte Biol 06 - Aging Micro Array

    5/14

    Bmp1) were also reduced in the aged, and IL-16 (Il16) and

    IL-1f6 (Il1f6) were increased (Table 1). IL-16 is known to

    promote proinflammatory cytokine production, but the mecha-

    nisms are not fully understood [28]. IL-1f6 is a new IL-1 family

    member, whose effects are not fully known but does not appear

    to replace the function of reduced IL-1 in the aged [29].

    Increased expression of these cytokines needs to be verifiedfurther by ELISA or RT-PCR. Many of the chemokines that

    were reduced in the aged are involved in innate immunity and

    inflammation. Thus, chemokines CCL4, CXCL1 (or keratino-

    cyte-derived chemokine), CCL6, CCL9, and CCL24 (or

    eotaxin-2; gene symbols are Ccl4, Cxcl1, Ccl6, Ccl9, and

    Ccl24), as well as the receptors CC chemokine receptor 3

    (CCR3) and CCR5 (Ccr3 and Ccr5), involved in chemotaxis of

    neutrophils, M, and eosinophils, are reduced in the aged

    (Table 1), which goes well with a reduction in the overall

    inflammatory response in spleens from the aged. CCBP2

    (Ccbp2), which is up-regulated in the aged, is a protein that has

    no signaling domain, suggesting that it could further inhibit

    chemokine function [30]. In addition, a variety of chemokines

    and receptors (CXCL9, CXCL10, CXCL11, CCR7), which af-

    fect CD4 and CD8 T cell migration and T helper cell type 1

    (Th1) development, are reduced in the aged. Reduction of some

    of these chemokines (IP-9, IP-10, monokine induced by IFN-)

    could be secondary to the reduction in IFN-, as they are allinduced by IFN-. This is in agreement with an age-associated

    decrease in T cell function and in particular, Th1 cell function.

    TLR signaling is reduced in the LPS-stimulatedM from the aged

    As stimulation with LPS led to a differential regulation of

    cytokine genes in M from aged and young mice, we decided

    to further scrutinize the TLR pathway and its regulation. Upon

    LPS stimulation, a majority of the genes in the TLR pathway

    had higher levels of expression in M from the young than

    those from the aged [e.g., TNF-receptor-associated factor 6

    Fig. 3. Microarray data validate previous data ob-

    tained by ELISA. The RNA extracted from M

    stimulated for 6 h with LPS was analyzed by mi-

    croarray; data are shown for pro- and anti-inflam-

    matory cytokines (left column). The corresponding

    amounts of IL-1, -6, -10, and -12 protein secreted

    into culture supernatants after stimulation with LPS

    for 1 day, as determined by ELISA, are shown in the

    right column. Means identified by the same symbol

    are statistically different (P

    0.05). The ELISAdata are representative of at least six independent

    experiments.

    1318 Journal of Leukocyte Biology Volume 79, June 2006 http://www.jleukbio.org

  • 8/6/2019 Chelvarajan - Leukocyte Biol 06 - Aging Micro Array

    6/14

    (Traf6), Cd14, Rel, and Relb; Table 2]. However, a relatively

    small group of TLR components was up-regulated considerablyin M from aged mice, resulting in higher levels of expression

    than in M from young (e.g., Mapk14). Table 2 contains a

    partial list of key components of the TLR pathway affected in

    M from the aged. The p100, p105, and p65 subunits of

    NF-B (Nfkb2, Nfkb1, and Rela) together with other compo-

    nents of the pathway were also reduced in M from the aged

    stimulated with LPS.

    As a defect in the TLR pathway could account for an

    alteration in not only a response to LPS but also to other TLR

    ligands, the expression of a number of key components of the

    TLR pathway was validated by RT-PCR. Thus, the reduction of

    myeloid differentiation primary-response protein 88 (MyD88)

    and TRAF-6 (Myd88 and Traf6), adaptor molecules that arecritical for activation of the NF-B pathway, in M from the

    aged was validated further by RT-PCR (Fig. 4 and Table 2). In

    addition to a decrease in the several components required for

    activation of the NF-B pathway, there was also an increase in

    a few components of the pathway, such as IRAK-M and TIRAP

    (Irak-3 and Tirap; Fig. 4 and Table 2). IRAK-M negatively

    regulates TLR signaling, and TIRAP is a critical upstream

    adaptor molecule for TLR2 responses, and its increased ex-

    pression may have a role in the altered response of M from

    the aged to LPS [31]. As these two genes did not fit the general

    pattern of TLR genes (i.e., reduction in M from the aged),

    TABLE 1. Expression of Key Cytokine and Chemokine Genes

    Gene title Symbol AliasAged LPS/Young LPS Pair-wise P

    Chemokine-binding protein 2 Ccbp2 24.5 6.E-05Chemokine (CC motif) ligand 24 Ccl24 Eotaxin-2 0.4 6.E-03Chemokine (CC motif) ligand 4 Ccl4 MIP-1 0.4 1.E-08Chemokine (CC motif) ligand 6 Ccl6 MRP-1 0.3 9.E-05Chemokine (CC motif) ligand 9 Ccl9 MIP-1 /MRP-2 0.3 1.E-09Chemokine (CC motif) receptor 3 Ccr3 MIP-1RL2 0.2 3.E-05Chemokine (CC motif) receptor 5 Ccr5 0.5 2.E-03Chemokine (CC motif) receptor 7 Ccr7 0.4 5.E-09Chemokine (CC motif) receptor-like 2 Ccrl2 0.4 9.E-09Colony-stimulating factor 1 (macrophage) Csf1 M-CSF 0.3 2.E-04Colony-stimulating factor 2 (granulocyte-macrophage) Csf2 GM-CSF 0.2 4.E-06Chemokine (CXC motif) ligand 1 Cxcl1 Gro1 0.5 6.E-05Chemokine (CXC motif) ligand 10 Cxcl10 IP-10 0.4 5.E-08Chemokine (CXC motif) ligand 11 Cxcl11 0.1 6.E-07Chemokine (CXC motif) ligand 16 Cxcl16 SR-PSOX 0.4 7.E-04Chemokine (CXC motif) ligand 9 Cxcl9 0.1 5.E-07Growth differentiation factor 15 Gdf15 MIC-1 0.4 2.E-06Interferon ( and ) receptor 1 Ifnar1 0.5 2.E-04Interferon ( and ) receptor 2 Ifnar2 0.5 1.E-06Interferon- Ifng 0.1 1.E-07Interleukin 1 Il1b IL-1 0.6 7.E-08

    Interleukin 1 receptor antagonist Il1rn IL-1RA 0.5 4.E-06Interleukin 12a Il12a 0.4 2.E-11Interleukin 13 receptor, 1 Il13ra1 0.4 1.E-03Interleukin 15 Il15 0.2 2.E-12Interleukin 15 receptor, chain Il15ra 0.2 2.E-09Interleukin 16 Il16 3.5 2.E-07Interleukin 1 Il1a 0.2 7.E-10Interleukin 1 family, member 6 Il1f6 2.5 3.E-08Interleukin 1 receptor-like 1 Il1rl1 0.5 3.E-03Interleukin 1 receptor antagonist Il1rn 0.4 4.E-06Interleukin 23, subunit p19 Il23a 0.5 2.E-05Interleukin 2 receptor, chain Il2ra 0.4 5.E-08Interleukin 4 Il4 0.2 7.E-06Interleukin 6 Il6 0.4 3.E-10Leukemia inhibitory factor Lif 0.3 4.E-05

    Transforming growth factor- 1 Tgfb1 TGF- 0.9 2.E-02Tumor necrosis factor Tnf 0.5 1.E-10Tumor necrosis factor receptor superfamily, member 1a Tnfrsf1a TNF-R1 0.5 5.E-06Tumor necrosis factor receptor superfamily, member 1b Tnfrsf1b TNF-R2 0.5 4.E-06Tumor necrosis factor (ligand) superfamily, member 14 Tnfsf14 LIGHT 0.3 3.E-04

    Data generated from microarray analysis. Fold-change represents the ratio of hybridization intensity of any gene in the aged to that in the young M treated

    with LPS. Only genes with a statistical significance at the level of P 0.01 are shown. Although we routinely used a fold-change filter of 2.0 (or 0.5), this table

    includes some biologically relevant genes that do not meet this criterion. MRP-1, Migration inhibitory factor-related protein-1; GM-CSF, granulocyte macrophage-

    colony stimulating factor; IP-10, interferon (IFN)-inducible protein 10; SR-PSOX, scavenger receptor for phosphatidylserine and oxidized lipoprotein; MIC-1,

    macrophage-inhibitory cytokine-1; IL-IRA, IL-1R agonist; TGF-, transforming growth factor-; LIGHT, homologous to lymphotoxin, exhibits inducible

    expression, competes with herpes virus glycoprotein D for herpes virus entry mediator on T cells.

    Chelvarajan et al. Macrophages from aged mice are defective and have increased p38 MAPK 1319

  • 8/6/2019 Chelvarajan - Leukocyte Biol 06 - Aging Micro Array

    7/14

    TABLE 2. Expression of Key Components of the TLR Pathway

    Gene title Symbol AliasAged LPS/Young LPS Pair-wise P

    Adaptors & TLR-interacting proteinsHeat shock protein 1A Hspa1a 0.3 8.E-07Lymphocyte antigen 96 Ly96 0.6 6.E-06Myeloid differentiation primary response gene 88 Myd88 0.6 9.E-09Pellino 1 Peli1 0.4 5.E-04Peptidoglycan recognition protein 1 Pglyrp1 4.3 4.E-08

    Receptor (TNFRSF)-interacting serine-threonine kinase 2 Ripk2 0.4 6.E-08Toll-like interleukin 1 receptor (TIR) domain containing adaptor protein Tirap 1.9 4.E-03

    EffectorsCaspase 8 Casp8 0.6 3.E-05Interleukin-1 receptor-associated kinase 1 Irak1 1.9 3.E-04Interleukin-1 receptor-associated kinase 2 Irak2 0.6 2.E-05Interleukin-1 receptor-associated kinase 3 Irak3 IRAK-M 2.1 2.E-06Mitogen-activated protein kinase kinase kinase 7-interacting protein 2 Map3k7ip2 Tab2 0.6 7.E-07TNF receptor-associated factor 6 Traf6 0.5 3.E-03

    IRF pathwayChemokine (CXC motif) ligand 10 Cxcl10 IP-10 0.4 5.E-08Interferon regulatory factor 1 Irf1 0.3 4.E-10Interferon regulatory factor 7 Irf7 0.4 4.E-06TANK-binding kinase 1 Tbk1 0.5 1.E-03

    MAPK pathwayMitogen-activated protein kinase kinase 4 Map2k4 MEK4 0.6 5.E-05Mitogen-activated protein kinase 14 Mapk14 p38 MAPK 2.9 1.E-03Mitogen-activated protein kinase 1 Mapk1 Erk-2 1.3 1.E-03Mitogen-activated protein kinase 3 Mapk3 Erk-1 1.4 3.E-06Mitogen-activated protein kinase 8 Mapk8 JNK1 1.1 3.E-01Mitogen-activated protein kinase 9 Mapk9 JNK2 0.8 1.E-03

    NF/IL-6 pathwayProstaglandin E synthase Ptges mPGES-1 0.4 1.E-06Prostaglandin-endoperoxide synthase 2 Ptgs2 Cox-2 0.3 4.E-08

    NF-B pathwayInhibitor of B kinase Ikbkg NEMO 0.6 3.E-03Interleukin-12a Il12a 0.4 2.E-11Mitogen-activated protein kinase kinase kinase 14 Map3k14 Nik 3.2 1.E-04Mitogen-activated protein kinase kinase kinase kinase 4 Map4k4 0.5 7.E-05Ubiquitin-conjugating enzyme E2D 3 (UBC4/5 homolog, yeast) Nfkb1 p50/p105 0.3 7.E-06Nuclear factor of light polypeptide gene enhancer in B cells 2, p49/p100 Nfkb2 p52 0.5 2.E-07Nuclear factor of light-chain gene enhancer in B cells inhibitor, Nfkbib I-B 0.5 5.E-10Nuclear factor of light polypeptide gene enhancer in B cells inhibitor, Nfkbie IKBE 0.4 2.E-08Reticuloendotheliosis oncogene Rel c-Rel 0.6 1.E-05v-Rel reticuloendotheliosis viral oncogene homolog A (avian) Rela p65 0.5 1.E-06Tumor necrosis factor Tnf TNF- 0.5 1.E-10Tumor necrosis factor receptor superfamily, member 1a Tnfrsf1a TNF-R1 0.5 5.E-06

    Regulation of adaptive immunity

    CD80 antigen Cd80 B7-1 0.6 3.E-03

    Toll-like receptorsLymphocyte antigen 64 Ly64 Muc13 0.5 1.E-03Toll-like receptor 2 Tlr2 0.4 1.E-07Toll-like receptor 4 Tlr4 1.0 8.E-01Toll-like receptor 6 Tlr6 0.6 5.E-08Toll-like receptor 9 Tlr9 1.0 9.E-01

    Data generated from microarray analysis. Fold-change represents the ratio of hybridization intensity of any gene in M from the aged to that in those from young

    mice treated with LPS. Only genes with a statistical significance at the level of P 0.01 are shown. Although we routinely used a fold-change filter of 2.0 (or

    0.5), this table includes some biologically relevant genes that do not meet this criterion. TNFRSF, TNF superfamily; Tab2, TGF -activated kinase-1-binding

    protein 2; IRF, IFN-regulatory factor; TANK, TRAF family member-associated nuclear factor (NF)-B activator; MEK4, MAPK kinase 4; MPGES-1, membrane

    PGE synthase-1; NEMO, NF-B essential modulator; NIK, NF-inducing kinase; 1-B, inhibitor of -B.

    1320 Journal of Leukocyte Biology Volume 79, June 2006 http://www.jleukbio.org

  • 8/6/2019 Chelvarajan - Leukocyte Biol 06 - Aging Micro Array

    8/14

    they were analyzed further by real-time RT-PCR, which vali-

    dated the microarray data (Fig. 4).The reduction in the com-

    ponents of NF-B signaling pathway explains the reduction in

    production of several proinflammatory cytokines and chemo-

    kines in LPS-stimulated M from the aged, as this pathway is

    known to be critical for the production of such soluble medi-

    ators. This is consistent with a global defect in the responses ofM from the aged, not only to LPS but also to TLR2 ligands

    such as peptidoglycan and S. pneumoniae bacteria and TLR9

    ligand CpG (unpublished observations).

    Effect of aging on the MAPK pathway

    LPS-induced cytokine secretion from M has been shown to

    be dependent on activation of the MAPKs: ERK1/2 (Mapk3

    and Mapk1), JNK1/2 (Mapk8 and Mapk9), and p38 MAPK

    (Mapk14). The microarray data did not find significant changes

    in the ERK and JNK levels, but the level of p38 MAPK

    mRNA was enhanced significantly in resting and LPS-stimu-

    lated M from the aged (Table 2 and Fig. 5A). To determine

    if the increased levels of mRNA reflect an increased level of

    p38 MAPK protein, a Western blot analysis was performed on

    lysates from M. The Western blot demonstrated that levels of

    total p38 MAPK protein (normalized to -actin levels to correct

    for differences in protein loading) were increased in M from

    the aged (Fig. 5, B and C). We then verified that M from theaged also had increased amounts of phospho-p38 MAPK, the

    functionally active form of p38 MAPK (Fig. 5, B and D). In this

    figure, phospho-p38 levels were normalized to -actin, which

    was similar in both age groups rather than to total p38 MAPK,

    as total p38 MAPK was different in the two age groups.

    The microarray study indicated that dual-specificity phospha-

    tase (DUSP)-10 (or MAPK-activating protein kinase 5; gene sym-

    bol is Dusp10), a newly discovered DUSP, was present in higher

    amounts in the M from the aged (data not shown). As a knockout

    of DUSP-10 had increased p38 MAPK activity [32], we verified by

    real-time RT-PCR that M from aged mice did indeed have an

    Fig. 4. Validation of microarray data by real-time

    RT-PCR. Multiple samples of mRNA, including

    those not analyzed by microarray, were transcribed

    into cDNA and analyzed by real-time PCR. RNA

    was extracted from three or more cultures of M

    and transcribed separately into cDNA. PCRs for

    Toll-IL-1R translation initiation region domain-

    containing adaptor protein (Tirap), Myd88, Irak3,

    and Traf6 were done in triplicate for each sample of

    cDNA. Intensities derived from each PCR were

    normalized to the corresponding values for 18SRNA, also obtained from real-time PCR. Means

    identified by the same symbol were statistically

    different (P0.05).

    Chelvarajan et al. Macrophages from aged mice are defective and have increased p38 MAPK 1321

  • 8/6/2019 Chelvarajan - Leukocyte Biol 06 - Aging Micro Array

    9/14

    increased expression ofDusp10 (data not shown). To determine if

    the increased phospho-p38 MAPK could play a role in the altered

    pattern of cytokine secretion of M from the aged SB203580, a

    well-characterized inhibitor to p38 MAPK was used. Inhibiting

    p38 MAPK in M from the aged resulted in reduced production

    of IL-10 (Fig. 6A). It is surprising that there was an increase in

    the production of IL-6, IL-12, and TNF- at low doses of

    SB203580 (Fig. 6, B and C, and data not shown). This increase

    was found in cultures of M from both age groups. The dose-

    response curve showed a steady inhibition of IL-10 synthesis withan increasing dose of SB203580, and TNF- and IL-12 were

    enhanced at low doses of the inhibitor but were suppressed at

    higher doses. This is consistent with the hypothesis that a thresh-

    old level of p38 MAPK is required for the synthesis of all cyto-

    kines, but after a certain level, it is inhibitory for the pro- but not

    anti-inflammatory cytokines.

    In contrast to p38 MAPK, which is elevated in the aged, our

    microarray data showed only a marginal change in ERK1/2

    mRNA (Table 2). Western blot analysis also showed that M

    from the aged had slightly reduced amounts of total protein

    levels of ERK1/2 (Fig. 7, A and B). However, when these

    blots were probed for phosphorylated forms of ERK, there was

    a dramatic reduction in phoshpho-ERK1/2 in M from the

    aged before and after stimulation with LPS (Fig. 7, A and C).

    This reduction in ERK may also contribute to the decreased

    cytokine production in the aged.

    DISCUSSION

    The primary goal of this microarray study was to understandthe molecular basis of cytokine dysregulation in M from aged

    mice, which was in part responsible for the decreased antibody

    response of aged mice to pneumococcal polysaccharides. The

    major findings from this analysis are that there is a more

    extensive dysregulation in cytokine production and TLR and

    MAPK signaling in M from the aged than previously appre-

    ciated and that the phenotype of M from the aged does not fit

    into the known patterns of M heterogeneity.

    As noted in previous studies, LPS stimulation not only

    induces expression of many genes but also represses many

    genes that are constitutively expressed in both age groups [25,

    Fig. 5. M from the aged have higher amounts of p38 MAPK. Data from microarray show that M from the aged have more p38 MAPK mRNA (A). M were rested

    at 37C for 90 min and then stimulated in duplicate with LPS for 15 min. The cells were lysed, subjected to SDS-PAGE, transferred to polyvinylidene difluoride, and then

    probed for phospho-p38 MAPK; stripped and then probed for total p38 MAPK; and stripped and then probed for -actin (B). Total p38 MAPK was normalized to -actin

    (C), and phosphorylated p38 MAPK was normalized to -actin (D). In this panel, the data points come from two different experiments, yielding a total of six values foreach treatment with the exception of M from the aged LPS, which only had four values. Means identified by the same symbol are statistically different (P0.05).

    1322 Journal of Leukocyte Biology Volume 79, June 2006 http://www.jleukbio.org

  • 8/6/2019 Chelvarajan - Leukocyte Biol 06 - Aging Micro Array

    10/14

    33]. It is surprising that the numbers of LPS-repressed genes in

    both age groups are rather large and greater than those induced

    by LPS. Some of these repressed genes include PPAR-,

    CCL24, and CCR1 (gene symbols are Ppar, Ccl24, and Ccr1,

    respectively), although these were suppressed by LPS to the

    same extent in both age groups. CCL24 (or eotaxin-2) is a

    chemokine involved in recruitment of eosinophils and ba-

    sophils to the sites of inflammation, and its suppression may

    control the type of inflammatory response to be induced. Sim-

    ilarly, PPAR- has been shown to inhibit production of several

    inflammatory mediators such as TNF-, IL-1, IL-6, and induc-

    ible nitric oxide synthase (NOS) in M, and its suppression by

    LPS may be a prerequisite for the induction of the LPS-induced

    inflammatory phenotype [34, 35]. However, the suppression of

    PPAR- was similar in both age groups, eliminating it as a

    possible candidate for the differential production of cytokines

    by M from the young and aged mice. Moreover, the microar-

    ray study has allowed us to eliminate the possibility that

    elevated production of inhibitory cytokines, including TGF-

    (Tgfb), suppressor of cytokine signaling family molecules, or

    IL-1RA (Il1rn; Table 1 and data not shown), is responsible for

    the anti-inflammatory phenotype in M from the aged, as none

    of these genes was expressed at elevated levels in M from

    aged in comparison with young mice [3638].

    M heterogeneity has been recognized recently, and an

    imbalance in M subsets could be a reason for the difference

    between the young adult versus the aged. First, M have been

    subdivided into M-1 and M-2 phenotypes depending on their

    ability to produce NO and proinflammatory cytokines (M-1type) or anti-inflammatory agents such as IL-1RA and arginase

    (M-2 type), suggesting a possibility that one of these types of

    M accumulates in the spleens of the aged [39]. Our gene

    expression analysis has shown this to be unlikely, as NOS-2

    and arginase, respectively, unique to M-1 and M-2 M, were

    reduced in M from the aged (data not shown). Second,

    resident alveolar M are known to be anti-inflammatory as a

    result of constitutive production of IL-10, but the effect of

    IL-10 is overcome by TLR agonists [40]. The splenic M from

    the aged are unlike the alveolar M from young adult mice, as

    they do not produce IL-10 constitutively, and TLR4 ligands do

    not overcome their defects in proinflammatory cytokine pro-duction. Third, they are also distinct from the anti-inflamma-

    tory M from the intestine, which neither express CD11b (and

    many other M cell surface receptors) nor produce IL-1,

    IL-10, and IL-12, whereas the M from the aged are

    CD11bve and produce IL-10 in excess [41]. Fourth, it has

    also been shown that M can be alternatively activated by

    IL-4, leading to suppression of proinflammatory cytokines and

    enhanced expression of major histocompatibility complex class

    II (MHC II) genes as well as IL-1RA [42]. As the aged have

    been shown to have an increased incidence of Th2 T cells [43],

    it was conceivable that the M in the aged have markers of

    IL-4 activation. Our cytokine expression pattern and the gene

    expression analysis have shown that splenic M from the ageddo not have this alternative activation phenotype (no increase

    in IL-1ra or MHC II; data not shown). Fifth, Mosser and

    colleagues [44, 45] have shown that LPS immune complexes

    can induce yet another activation pattern, resulting in in-

    creased IL-10 and decreased production of IL-12 with no effect

    on TNF- production. As the aged have been shown to have

    increased autoantibodies, it is plausible that M from the aged

    are responding as if they have encountered immune complexes.

    This is unlikely, as our microarray study has shown that M

    from the aged exhibit decreased expression of most proinflam-

    matory cytokines and chemokines, such as Il1b, Il6, and Tnf

    Fig. 6. Inhibiting p38 MAPK enhances production of proinflammatory cyto-

    kines and suppresses production of anti-inflammatory cytokines. M were

    cultured in duplicate with LPS and various amounts of S203580, a p38 MAPKinhibitor, for 24 h. The supernatant was then assayed in triplicate by ELISA for

    IL-10 (A), TNF- (B), and IL-12 (C). Students t-test was performed with

    means identified by the same symbol. These data are representative of three

    independent experiments.

    Chelvarajan et al. Macrophages from aged mice are defective and have increased p38 MAPK 1323

  • 8/6/2019 Chelvarajan - Leukocyte Biol 06 - Aging Micro Array

    11/14

    (Table 1). Thus, the studies from our laboratory have identified

    a uniquely hyporesponsive M in spleens from the aged,

    which has profound influences on immune responses to poly-saccharide antigens and may affect the overall ability of the

    aged to generate an inflammatory response necessary to contain

    infections.

    Although the response of M from the aged to LPS (i.e., a

    TLR ligand) is altered significantly, the expression of several

    TLR members (Tlr4, Tlr6, and Tlr9), with the exception ofTlr2,

    is comparable with that of M from the young (Table 2). This

    finding is consistent with previous reports from Boehmer et al.

    [20, 21], who also found no decrease in these receptors, but

    disagrees with that of Renshaw et al. [15]. However, our finding

    that the downstream signaling components, such as the adaptor

    molecule MyD88, and several members of the NF-B pathway,

    such as Rel-a, Rel-b, NF-B p50 and p52, and TRAF6(Myd88, Rela, Relb, Nfkb1, Nfkb2, and Traf6), were also re-

    duced in the aged suggests that the TLR-dependent pathway is

    working at a significantly reduced efficiency. It is interesting

    that the NF-B-independent pathway is also reduced, as com-

    ponents of this pathway, such as TANK-binding kinase 1 and

    IRF-1 (Tbk1 and Irf1), are reduced in LPS-stimulated M

    (Table 2). Superimposed on the reduction of these TLR signal-

    ing pathway intermediates needed for positive signaling, levels

    of IRAK-M (Irak3), a known negative regulator of this pathway,

    are enhanced in M from the aged. Thus, there is an overall

    reduction in the TLR signaling pathway, which may account for

    the generalized decrease in the proinflammatory cytokine se-

    cretion from M from the aged. Presently, it is unclear why so

    many components of the TLR signaling pathway intermediatesare reduced in the aged (or increased in the case of the

    negative regulator IRAK-M), as they are not known to be on the

    same chromosome, ruling out a coordinated regulation of sev-

    eral of these genes in the aged.

    The most surprising finding of our microarray study is the

    significant increase in the levels of p38 MAPK in M from

    the aged, independent of LPS stimulation. It is of interest that

    age-associated changes in the mRNA levels for other major

    MAPKs, such as ERK1/2 and JNK1/2, were minimal or un-

    changed. This was confirmed at the protein level for p38

    MAPK and ERK1/2 by Western blot analysis (Figs. 5 and 7).

    Not only was there an increase in the total p38 MAPK level,

    but the level of the functionally active, phosphorylated form ofthe enzyme was also elevated in M from the aged before and

    after stimulation with LPS. It is notable that Iwasa et al. [46]

    found that senescent fibroblasts express higher levels of acti-

    vated p38 MAPK, and this elevated phospho-p38 MAPK has a

    causal role in the senescent phenotype of the fibroblast cells.

    Thus, inhibition of the p38 MAPK activity enhanced the pro-

    liferation capability of fibroblasts, whereas expression of con-

    stitutively active MEK (MKK)6, an activator of p38 MAPK,

    induced a senescent phenotype in fibroblasts from young [46].

    Our findings about an increase in p38 MAPK in the aged M

    are in contrast to the results of Boehmer et al. [20, 21], who

    Fig. 7. M from the aged have dramatically reduced

    amounts of phospho-ERK1/2. The lysates used in Figure

    5 were also probed for total ERK1/2 and phospho-

    ERK1/2 (A). (B) Amount of total ERK1/2 was normalized

    to -actin. For this determination, duplicate cultures

    were pooled. (C) The amounts of phospho-ERK1/2 nor-

    malized to total ERK1/2 are shown. Means identified by

    the same symbol are statistically different (P0.05).

    These data are representative of two independent exper-

    iments.

    1324 Journal of Leukocyte Biology Volume 79, June 2006 http://www.jleukbio.org

  • 8/6/2019 Chelvarajan - Leukocyte Biol 06 - Aging Micro Array

    12/14

    found a decrease in the total and phosphorylated forms of p38

    MAPK. The discrepancy could be a result of the use of thio-

    glycollate-induced peritoneal M in one study by these au-

    thors [20]. Although splenic M were examined in the second

    study, the Western blots in both studies were not normalized to

    a housekeeping protein such as actin for equal loading. This

    may affect the interpretation of the data.

    Consistent with the published reports about the need for p38

    MAPK for IL-10 gene expression, inhibition of p38 MAPK

    activity with SB203580 led to a dose-dependent inhibition of

    LPS-induced IL-10 production in both age groups. However,

    p38 MAPK has also been implicated in the production of

    several proinflammatory cytokines, such as TNF-, IL-1, and

    IL-6 in numerous studies and in many different cell types such

    as monocytes, M, and dendritic cells (DC) [47, 48]. More-

    over, inhibition of p38 MAPK reduced inflammation and sepsis

    in some animal models [49, 50]. In contrast, Li et al. [51]

    recently reported that p38 MAPK is crucially involved in

    osteoclast production but not cytokine production by bone

    marrow-derived M. It turns out that most studies that use

    p38-specific inhibitors in vitro have used various cell lines

    such as RAW264.7, THP-1, and 70Z/3 transfected with CD14,

    and few of them have performed detailed, dose-response stud-ies. Although the in vivo data clearly establish the anti-inflam-

    matory effects of the p38 MAPK inhibitors, it is difficult to

    know the critical cell that is affected. Thus, our data, showing

    that at low doses, the p38 MAPK inhibitor SB203580 enhances

    production of the proinflammatory cytokines TNF-, IL-12,

    and IL-6 (Fig. 6 and data not shown), are rather unique and

    ascribe a negative and a positive role for p38 MAPK in causing

    an inflammatory phenotype. This dose response may explain

    why in the literature, there are conflicting reports about the

    requirement of p38 MAPK for the synthesis of pro- and anti-

    inflammatory cytokines [44, 52, 53].

    The ability of low doses of the p38 inhibitor to enhanceproinflammatory cytokines is consistent with our finding that

    total and phospho-p38 levels are enhanced in the aged. Our

    data, suggesting that at higher doses, the p38 MAPK inhibitor

    reduces proinflammatory cytokines, are consistent with the

    published literature. We hypothesize that certain minimal lev-

    els of this enzyme are required for production of these cyto-

    kines, but at higher levels, p38 MAPK may actually inhibit

    production of IL-6 and IL-12. As it is well known that the

    expression of many cytokine genes (TNF- and IL-1 among

    others) is regulated at the transcriptional level and at the level

    of mRNA stability, it is conceivable that the low and high

    concentrations of active phospho-p38 MAPK influence these

    two processes differently [50, 52, 54]. In contrast, p38 MAPKdoes not appear to have any negative effect on IL-10 produc-

    tion, as IL-10 levels were decreased in a dose-dependent

    manner with the p38 inhibitor.

    We have shown previously that the altered pattern of cyto-

    kine production in M from aged mice was a result of an

    excess production of IL-10 [1]. The neutralization of IL-10

    resulted in enhanced production of proinflammatory cytokines,

    to levels comparable with control M from young mice. In this

    study, we see that by partially suppressing p38 MAPK activity

    in M from aged mice, a reduction in IL-10 occurred, with a

    concomitant enhancement of proinflammatory cytokine produc-

    tion, similar to what was seen when IL-10 was neutralized [1].

    This would lead us to postulate that the increased level of

    LPS-induced IL-10 seen in M from aged mice is a result of

    the higher amount of p38 MAPK activity in M from aged

    mice.

    Unlike p38 MAPK, levels of ERK1/2 were similar in M

    from both age groups. However, levels of phosphorylated

    ERK1/2 were reduced significantly in M from the aged. ERK

    has also been shown to be important for LPS-induced secretion

    of cytokines such as IL-1, IL-6, and TNF- and for LPS

    immune complexes, induced production of IL-10 from M [44,

    55, 56]. Conversely, Dillon et al. [57] found that ERK activa-

    tion inhibits production of IL-12 by inducing c-fos in DC and

    that c-fos-negative DC have elevated levels of IL-12 [58]. A

    negative role for ERK in LPS-induced M production of IL-12

    has not been described so far. Data presented here suggest that

    ERK may not be having such a negative role in production of

    IL-1, IL-6, IL-12, or TNF-, as all of these cytokines are

    reduced in M from the aged, which have dramatically re-

    duced levels of active ERK. Presently, we have not tested if

    ERK has a negative role in IL-10 production. Our results

    suggest that a balance between functionally active ERK and

    p38 MAPK is required for a pattern of cytokine productionsuch as that seen in LPS-stimulated M from young mice and

    that a loss of this balance leads to the cytokine-dysregulated

    phenotype of the aged. To further clarify the mechanism of

    age-associated cytokine dysregulation, we will also be assess-

    ing the levels of functionally active JNK in M from the aged,

    as JNK has been shown to have positive and negative roles in

    cytokine secretion by M [47, 59]. Another question that

    remains is about the mechanisms that are responsible for p38

    MAPK up-regulation in M from the aged, as in most inflam-

    matory responses, the levels of these enzymes are not changed,

    but their functional activities are regulated by various stimuli

    [47].In addition to the direct effects of aging on p38 MAPK and

    ERK expression as well as activation, some of the signaling

    proteins that regulate the MAPK pathway are also affected by

    aging. Thus DUSP-10, an enzyme known to inhibit MAPK

    signaling, is elevated in aged M before and after LPS stim-

    ulation in comparison with the young M. Mice in which

    DUSP-10 is deleted have an increase in the production of

    proinflammatory cytokines, in part, as a result of an increase in

    activities of p38 and JNK MAPK, but the cellular source of

    these cytokines was not identified [32]. Presently, we are

    investigating the contribution of DUSP-10 to the cytokine-

    dysregulated phenotype of the aged M.

    In summary, our microarray analysis has shown that Mfrom aged mice have a global defect in the TLR signaling

    pathway and in production of proinflammatory cytokines and

    chemokines, and the anti-inflammatory cytokines are in-

    creased, such that the splenic M in the aged have an anti-

    inflammatory phenotype. We find that the aged mouse M

    have a unique phenotype, which is distinct from the currently

    known modes of M regulation. The aging signature for M

    includes an elevation of proliferation-specific genes. Our pre-

    liminary, immunohistochemistry studies with bromodeoxyuri-

    dine labeling show that indeed, there are more proliferating

    cells in the spleens of aged mice. Currently, we are investigat-

    Chelvarajan et al. Macrophages from aged mice are defective and have increased p38 MAPK 1325

  • 8/6/2019 Chelvarajan - Leukocyte Biol 06 - Aging Micro Array

    13/14

    ing the relation between this proliferation phenotype and the

    cytokine dysregulation phenotype in M from the aged mice.

    Finally, we have shown that the cytokine dysregulation is a

    result of an imbalance in MAPK activation (increased p38

    MAPK) and that inhibition of p38 MAPK partially restores

    production of cytokines such as IL-6 and IL-12 in M from the

    agedcytokines that are important for B cell responses.

    ACKNOWLEDGMENTS

    This work was supported in part by NIH Grants AG05731 and

    CA 92372 to S. B., AG-16824 to T. V. G., and P20-RR16481

    to A. J. S. Supplementary data for this article are available at

    National Cancer Institutes caArray data portal (http://

    caarray.nci.nih.gov). Our thanks are to Dr. Alan Kaplan

    for critical reading of this manuscript, Ms. Radhika Vaishnav

    for her help with developing the protocol for RNA extraction,

    and to Ms. Donna Wall and Dr. Kuey-Chu Chen for their expert

    help with the microarray analysis.

    REFERENCES

    1. Miller, R. A. (1996) The aging immune system: primer and prospectus.Science 273, 7074.

    2. Effros, R.B. (2001) Ageing and the immune system. Novartis Found.Symp. 235, 130139.

    3. Effros, R. B., Walford, R. L. (1984) The effect of age on the antigen-presenting mechanism in limiting dilution precursor cell frequency anal-ysis. Cell. Immunol. 88, 531539.

    4. Butler, J. C., Shapiro, E. D., Carlone, G. M. (1999) Pneumococcal vac-cines: history, current status, and future directions. Am. J. Med. 107,69S76S.

    5. Chelvarajan, R. L., Collins, S. M., Van Willigen, J. M., Bondada, S. (2005)The unresponsiveness of aged mice to polysaccharide antigens is a resultof a defect in macrophage function. J. Leukoc. Biol. 77, 503512.

    6. Artz, A. S., Ershler, W. B., Longo, D. L. (2003) Pneumococcal vaccinationand revaccination of older adults. Clin. Microbiol. Rev. 16, 308318.

    7. Landers, C. D., Chelvarajan, R. L., Bondada, S. (2005) The role of B cellsand accessory cells in the neonatal response to TI-2 antigens. Immunol.Res. 31, 2536.

    8. Chelvarajan, R. L., Collins, S., Doubinskaia, I. E., Goes, S., Van Willigen,J., Flanagan, D., De Villiers, W. J., Bryson, J. S., Bondada, S. (2004)Defective macrophage function in neonates and its impact on unrespon-siveness of neonates to polysaccharide antigens. J. Leukoc. Biol. 75,982994.

    9. Bondada, S., Wu, H., Robertson, D. A., Chelvarajan, R. L. (2000) Acces-sory cell defect in unresponsiveness of neonates and aged to polysaccha-ride vaccines. Vaccine 19, 557565.

    10. Bondada, S., Garg, M. (1994) Thymus independent antigens. In Handbookof B and T Lymphocytes (E. C. Snow, ed.), New York, NY, Academic,

    343370.11. Bohnsack, J. F., Brown, E. J. (1986) The role of spleen in resistance to

    infection. Annu. Rev. Med. 37, 4959.12. Kang, Y-S., Kim, J. Y., Bruening, S. A., Pack, M., Charalambous, A.,

    Pritsker, A., Moran, T. M., Loeffler, J. M., Steinman, R. M., Park, C. G.(2004) The C-type lectin SIGN-R1 mediates uptake of the capsularpolysaccharide ofStreptococcus pneumoniae in the marginal zone of mousespleen. Proc. Natl. Acad. Sci. USA 101, 215220.

    13. Kruetzmann, S., Rosado, M. M., Weber, H., Germing, U., Tournilhac, O.,Peter, H-H., Berner, R., Peters, A., Boehm, T., Plebani, A., Quinti, I.,Carsetti, R. (2003) Human immunoglobulin M memory B cells controllingStreptococcus pneumoniae infections are generated in the spleen. J. Exp.Med. 197, 939945.

    14. Stout, R. D., Suttles, J. (2005) Immunosenescence and macrophage func-tional plasticity: dysregulation of macrophage function by age-associatedmicroenvironmental changes. Immunol. Rev. 205, 6071.

    15. Renshaw, M., Rockwell, J., Engleman, C., Gewirtz, A., Katz, J., Sambhara,S. (2002) Impaired Toll-like receptor expression and function in aging.J. Immunol. 169, 46974701.

    16. Plowden, J., Renshaw-Hoelscher, M., Engleman, C., Katz, J., Sambhara, S.(2004) Innate immunity in aging: impact on macrophage function. AgingCell 3, 161167.

    17. Plackett, T. P., Boehmer, E. D., Faunce, D. E., Kovacs, E. J. (2004) Agingand innate immune cells. J. Leukoc. Biol. 76, 291299.

    18. Renshaw, B. R., Fanslow III, W. C., Armitage, R. J., Campbell, K. A.,Wright, B., Davison, B., Maliszewski, C. R. (1994) Humoral responses inCD40 ligand-deficient mice. J. Exp. Med. 180, 18891900.

    19. Meydani, S. N., Han, S. N., Wu, D. (2005) Vitamin E and immuneresponse in the aged: molecular mechanisms and clinical implications.

    Immunol. Rev. 205, 269284.20. Boehmer, E. D., Goral, J., Faunce, D. E., Kovacs, E. J. (2004) Age-

    dependent decrease in Toll-like receptor 4-mediated proinflammatorycytokine production and mitogen-activated protein kinase expression.J. Leukoc. Biol. 75, 342349.

    21. Boehmer, E. D., Meehan, M. J., Cutro, B. T., Kovacs, E. J. (2005) Agingnegatively skews macrophage TLR2- and TLR4-mediated pro-inflamma-tory responses without affecting the IL-2-stimulated pathway. Mech. Age-ing Dev. 126, 13051313.

    22. Nau, G. J., Richmond, J. F. L., Schlesinger, A., Jennings, E. G., Lander,E. S., Young, R. A. (2002) Human macrophage activation programsinduced by bacterial pathogens. Proc. Natl. Acad. Sci. USA 99, 15031508.

    23. Jiang, H., Van de Ven, C., Satwani, P., Baxi, L. V., Cairo, M. S. (2004)Differential gene expression patterns by oligonucleotide microarray ofbasal versus lipopolysaccharide-activated monocytes from cord blood ver-

    sus adult peripheral blood. J. Immunol. 172, 58705879.24. Lang, R., Patel, D., Morris, J. J., Rutschman, R. L., Murray, P. J. (2002)

    Shaping gene expression in activated and resting primary macrophages byIL-10. J. Immunol. 169, 22532263.

    25. Wells, C. A., Ravasi, T., Faulkner, G., Carninci, P., Okazaki, Y., Hayash-izaki, Y., Sweet, M., Wainwright, B., Hume, D. (2003) Genetic control ofthe innate immune response. BMC Immunol. 4, 5.

    26. Wells, C. A., Ravasi, T., Sultana, R., Yagi, K., Carninci, P., Bono, H.,Faulkner, G., Okazaki, Y., Quackenbush, J., Hume, D. A., Lyons, P. A.(2003) Continued discovery of transcriptional units expressed in cells ofthe mouse mononuclear phagocyte lineage. Genome Res. 13, 13601365.

    27. Hosack, D. A., Dennis Jr., G., Sherman, B. T., Lane, H. C., Lempicki,R. A. (2003) Identifying biological themes within lists of genes with EASE.Genome Biol. 4, R70.

    28. Wilson, K. C., Center, D. M., Cruikshank, W. W. (2004) The effect ofinterleukin-16 and its precursor on T lymphocyte activation and growth.

    Growth Factors 22, 97104.29. Towne, J. E., Garka, K. E., Renshaw, B. R., Virca, G. D., Sims, J. E.

    (2004) Interleukin (IL)-1F6, IL-1F8, and IL-1F9 signal through IL-1Rrp2and IL-1RAcP to activate the pathway leading to NF-{}B and MAPKs.J. Biol. Chem. 279, 1367713688.

    30. Jamieson, T., Cook, D. N., Nibbs, R. J. B., Rot, A., Nixon, C., McLean, P.,Alcami, A., Lira, S. A., Wiekowski, M., Graham, G. J. (2005) Thechemokine receptor D6 limits the inflammatory response in vivo. Nat.Immunol. 6, 403411.

    31. Beutler, B. (2004) Inferences, questions and possibilities in Toll-likereceptor signaling. Nature 430, 257263.

    32. Zhang, Y., Blattman, J. N., Kennedy, N. J., Duong, J., Nguyen, T., Wang,Y., Davis, R. J., Greenberg, P. D., Flavell, R. A., Dong, C. (2004)Regulation of innate and adaptive immune responses by MAP kinasephosphatase 5. Nature 430, 793797.

    33. Gao, J. J., Diesl, V., Wittmann, T., Morrison, D. C., Ryan, J. L., Vogel,

    S. N., Follettie, M. T. (2002) Regulation of gene expression in mousemacrophages stimulated with bacterial CpG-DNA and lipopolysaccharide.J. Leukoc. Biol. 72, 12341245.

    34. Clark, R. B. (2002) The role of PPARs in inflammation and immunity.J. Leukoc. Biol. 71, 388400.

    35. Welch, J. S., Ricote, M., Akiyama, T. E., Gonzalez, F. J., Glass, C. K.(2003) PPAR{} and PPAR{} negatively regulate specific subsets oflipopolysaccharide and IFN-{} target genes in macrophages. Proc. Natl. Acad. Sci. USA 100, 67126717.

    36. Starr, R., Willson, T. A., Viney, E. M., Murray, L. J., Rayner, J. R.,Jenkins, B. J., Gonda, T. J., Alexander, W. S., Metcalf, D., Nicola, N. A.,Hilton, D. J. (1997) A family of cytokine-inducible inhibitors of signaling.Nature 387, 917921.

    37. Donnelly, R. P., Dickensheets, H., Finbloom, D. S. (1999) The interleu-kin-10 signal transduction pathway and regulation of gene expression inmononuclear phagocytes. J. Interferon Cytokine Res. 19, 563573.

    1326 Journal of Leukocyte Biology Volume 79, June 2006 http://www.jleukbio.org

  • 8/6/2019 Chelvarajan - Leukocyte Biol 06 - Aging Micro Array

    14/14

    38. Elgert, K. D., Alleva, D. G., Mullins, D. W. (1998) Tumor-inducedimmune dysfunction: the macrophage connection. J. Leukoc. Biol. 64,275290.

    39. Mills, C. D., Kincaid, K., Alt, J. M., Heilman, M. J., Hill, A. M. (2000)M-1/M-2 macrophages and the Th1/Th2 paradigm. J. Immunol. 164,61666173.

    40. Fernandez, S., Jose, P., Avdiushko, M. G., Kaplan, A. M., Cohen, D. A.(2004) Inhibition of IL-10 receptor function in alveolar macrophages byToll-like receptor agonists. J. Immunol. 172, 26132620.

    41. Smythies, L. E., Sellers, M., Clements, R. H., Mosteller-Barnum, M.,Meng, G., Benjamin, W. H., Orenstein, J. M., Smith, P. D. (2005) Humanintestinal macrophages display profound inflammatory anergy despite avidphagocytic and bacteriocidal activity. J. Clin. Invest. 115, 6675.

    42. Gordon, S. (2003) Alternative activation of macrophages. Nat. Rev. Im-munol. 3, 2335.

    43. Hsu, H. C., Scott, D. K., Mountz, J. D. (2005) Impaired apoptosis andimmune senescencecause or effect? Immunol. Rev. 205, 130146.

    44. Lucas, M., Zhang, X., Prasanna, V., Mosser, D. M. (2005) ERK activationfollowing macrophage Fc{}R ligation leads to chromatin modifications atthe IL-10 locus. J. Immunol. 175, 469477.

    45. Anderson, C. F., Mosser, D. M. (2002) Cutting edge: biasing immuneresponses by directing antigen to macrophage Fc receptors. J. Immunol.168, 36973701.

    46. Iwasa, H., Han, J., Ishikawa, F. (2003) Mitogen-activated protein kinasep38 defines the common senescence-signaling pathway. Genes Cells 8,131144.

    47. Kyriakis, J. M., Avruch, J. (1996) Sounding the alarm: protein kinasecascades activated by stress and inflammation. J. Biol. Chem. 271,2431324316.

    48. Davis, R. J. (2000) Signal transduction by the JNK group of MAP kinases.Cell 103, 239252.

    49. Ono, K., Han, J. (2000) The p38 signal transduction pathway: activationand function. Cell. Signal. 12, 113.

    50. Roux, P. P., Blenis, J. (2004) ERK and p38 MAPK-activated proteinkinases: a family of protein kinases with diverse biological functions. Microbiol. Mol. Biol. Rev. 68, 320344.

    51. Li, X., Udagawa, N., Takami, M., Sato, N., Kobayashi, Y., Takahashi, N.(2003) p38 Mitogen-activated protein kinase is crucially involved inosteoclast differentiation but not in cytokine production, phagocytosis, ordendritic cell differentiation of bone marrow macrophages. Endocrinology144, 49995005.

    52. Lee, J. C., Young, P. R. (1996) Role of CSB/p38/RK stress responsekinase in LPS and cytokine signaling mechanisms. J. Leukoc. Biol. 59,152157.

    53. Mathur, R. K., Awasthi, A., Wadhone, P., Ramanamurthy, B., Saha, B.(2004) Reciprocal CD40 signals through p38MAPK and ERK-1/2 inducecounteracting immune responses. Nat. Med. 10, 540544.

    54. Rao, K. M. K. (2001) MAP kinase activation in macrophages. J. Leukoc.Biol. 69, 310.

    55. Rawadi, G., Ramez, V., Lemercier, B., Roman-Roman, S. (1998) Activa-tion of mitogen-activated protein kinase pathways by Mycoplasma fermen-tans membrane lipoproteins in murine macrophages: involvement in cy-tokine synthesis. J. Immunol. 160, 13301339.

    56. Valledor, A. F., Comalada, M., Xaus, J., Celada, A. (2000) The differentialtime-course of extracellular-regulated kinase activity correlates with themacrophage response toward proliferation or activation. J. Biol. Chem.275, 74037409.

    57. Dillon, S., Agrawal, A., Van Dyke, T., Landreth, G., McCauley, L., Koh,A., Maliszewski, C., Akira, S., Pulendran, B. (2004) A Toll-like receptor2 ligand stimulates Th2 responses in vivo, via induction of extracellularsignal-regulated kinase mitogen-activated protein kinase and c-Fos indendritic cells. J. Immunol. 172, 47334743.

    58. Agrawal, S., Agrawal, A., Doughty, B., Gerwitz, A., Blenis, J., Van Dyke,T., Pulendran, B. (2003) Cutting edge: different Toll-like receptor agonistsinstruct dendritic cells to induce distinct Th responses via differentialmodulation of extracellular signal-regulated kinase-mitogen-activated pro-tein kinase and c-Fos. J. Immunol. 171, 49844989.

    59. Utsugi, M., Dobashi, K., Ishizuka, T., Endou, K., Hamuro, J., Murata, Y.,Nakazawa, T., Mori, M. (2003) c-Jun N-terminal kinase negatively regu-lates lipopolysaccharide-induced IL-12 production in human macro-phages: role of mitogen-activated protein kinase in glutathione redoxregulation of IL-12 production. J. Immunol. 171, 628635.

    Chelvarajan et al. Macrophages from aged mice are defective and have increased p38 MAPK 1327