Skip to main content
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Eukaryotic Cell
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems
  • Log in
  • My alerts
  • My Cart

Main menu

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • Archive
    • Minireviews
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About IAI
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Eukaryotic Cell
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems

User menu

  • Log in
  • My alerts
  • My Cart

Search

  • Advanced search
Infection and Immunity
publisher-logosite-logo

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • Archive
    • Minireviews
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About IAI
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
Microbial Immunity and Vaccines

Inhibition of Major Histocompatibility Complex II Expression and Antigen Processing in Murine Alveolar Macrophages by Mycobacterium bovis BCG and the 19-Kilodalton Mycobacterial Lipoprotein

Scott A. Fulton, Scott M. Reba, Rish K. Pai, Meghan Pennini, Martha Torres, Clifford V. Harding, W. Henry Boom
Scott A. Fulton
1Division of Infectious Diseases
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Scott M. Reba
1Division of Infectious Diseases
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Rish K. Pai
2Institute of Pathology, Case Western Reserve University and University Hospitals of Cleveland, Cleveland, Ohio 44106-4984
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Meghan Pennini
2Institute of Pathology, Case Western Reserve University and University Hospitals of Cleveland, Cleveland, Ohio 44106-4984
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Martha Torres
1Division of Infectious Diseases
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Clifford V. Harding
2Institute of Pathology, Case Western Reserve University and University Hospitals of Cleveland, Cleveland, Ohio 44106-4984
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
W. Henry Boom
1Division of Infectious Diseases
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: whb@po.cwru.edu
DOI: 10.1128/IAI.72.4.2101-2110.2004
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

ABSTRACT

Alveolar macrophages constitute a primary defense against Mycobacterium tuberculosis, but they are unable to control M. tuberculosis without acquired T-cell immunity. This study determined the antigen-presenting cell function of murine alveolar macrophages and the ability of the model mycobacterium, Mycobacterium bovis BCG, to modulate it. The majority (80 to 85%) of alveolar macrophages expressed both CD80 (B7.1) and CD11c, and 20 to 30% coexpressed major histocompatibility complex II (MHC-II). Gamma interferon (IFN-γ) enhanced MHC-II but not B7.1 expression. Naive or IFN-γ-treated alveolar macrophages did not express CD86 (B7.2), CD11b, Mac-3, CD40, or F4/80. M. bovis BCG and the 19-kDa mycobacterial lipoprotein inhibited IFN-γ-regulated MHC-II expression on alveolar macrophages, and inhibition was dependent on Toll-like receptor 2. The inhibition of MHC-II expression by the 19-kDa lipoprotein was associated with decreased presentation of soluble antigen to T cells. Thus, susceptibility to tuberculosis may result from the ability of mycobacteria to interfere with MHC-II expression and antigen presentation by alveolar macrophages.

Experimental studies of granuloma formation and antimycobacterial immunity suggest that the lung is uniquely susceptible to Mycobacterium tuberculosis infection. Growth of virulent M. tuberculosis strains and avirulent strains of Mycobacterium bovis BCG is more rapid and destructive in the lung compared to other organs, and far fewer organisms are needed for infection when bacteria are delivered by aerosol compared to intravenous infection (9). Thus, infection of the lung is an important factor affecting the growth and survival of mycobacteria (4). The inhalation of aerosol droplets containing bacilli leads to the deposition of bacilli in both conducting and distal airways where many organisms are removed by mucociliary mechanisms. However, in alveolar spaces resident macrophages phagocytose organisms, resulting in the expression of reactive oxygen and nitrogen radicals and multiple chemokines and cytokines (3, 38, 39, 45, 50, 52). These innate immune responses do not control the early growth of either virulent M. tuberculosis or avirulent M. bovis BCG in murine lungs. Ultimately, control of both M. tuberculosis and M. bovis BCG infection is dependent on the recruitment and activation of major histocompatibility complex (MHC)-restricted CD4+ and CD8+ T cells (6, 9, 11, 21, 33, 52).

Whether permissive growth of M. tuberculosis or M. bovis BCG in alveolar spaces is due to an inability of alveolar macrophages to activate T cells, to the capacity of mycobacteria to impair alveolar macrophage function, or to the effects of alveolar protein factors on alveolar macrophage and T-cell function is not known. Recent studies have shown that the 19-kDa lipoprotein which is expressed by both M. bovis BCG and M. tuberculosis can inhibit MHC-II antigen processing and presentation in bone marrow-derived macrophages (31, 32, 34, 47). The inhibition occurs by blocking gamma interferon (IFN-γ) signaling through a Toll-like receptor 2 (TLR-2)-dependent mechanism. The present study was undertaken to determine the ability of alveolar macrophages to serve as antigen-presenting cells (APC) and whether alveolar macrophage function is affected by mycobacterial infection and exposure to mycobacterial lipoproteins. We determined that resident murine alveolar macrophages have the necessary surface molecules to serve as efficient APC for CD4+ T cells, that they can process and present MHC-II-restricted antigens, and that the APC function of alveolar macrophages was inhibited by M. bovis BCG infection and the 19-kDa lipoprotein. Thus, the mycobacterial infection of alveolar macrophages may impede activation of CD4+ T cells and contribute to the permissive pulmonary microenvironment supporting mycobacterial growth and persistence.

MATERIALS AND METHODS

Mice.Specific pathogen-free, female C57BL/6 (H-2b) mice were purchased from Charles River Laboratories (North Wilmington, Mass.) and used at between 8 and 12 weeks of age. TLR-2 gene knockout mice were generously provided by O. Takeuchi and S. Akira (Osaka University, Osaka, Japan) and bred onto the C57BL/6 background. Mice were housed under specific-pathogen-free conditions by using microisolator cages (Lab Products, Inc., Maywood, N.J.) and fed a standard rodent diet and water ad libitum. Studies were approved by the Institutional Animal Care and Use Committee at Case Western Reserve University.

Bacteria. M. bovis BCG Connaught (ATCC 35745) was grown at 37°C for 18 to 21 days in Proskeur-Beck medium (pH 7.0) containing l-asparagine (5.0 g/liter), potassium dihydrogen phosphate (5 g/liter), sesquimagnesium citrate (1.5 g/liter), dipotassium sulfate (0.5 g/liter), d-glucose (30 g/liter), and Tween-80 (0.05%). During mid-log growth, the culture was supplemented with glycerol (6%, vol/vol), aliquoted, and stored at −70°C until further use. Bacterial titers were determined by plating serial 10-fold dilutions on 7H10 agar and counting CFU after incubation for 2 to 3 weeks. The viability of M. bovis BCG is retained for at least 2 years. At the time of use, bacteria were thawed, sonicated for 40 s by means of an ultrasonic Cup Horn water bath (90 W; 20 kHz; Heat Systems-Ultrasonics, Farmingdale, N.Y.) and added to cultured alveolar macrophages as indicated.

Fluorescein labeling of M. bovis BCG.A stock solution of FLUOS [5(6)-carboxyfluorescein-N-hydroxysuccinimide ester] (Boehringer Mannheim, Mannheim, Germany) at a concentration of 20 mg/ml was made in dimethyl sulfoxide and stored at −20°C. A 1.0-ml aliquot of M. bovis BCG (3 × 107 to 5 × 107 CFU) was thawed, centrifuged at 4°C (10,000 × g for 15 min), resuspended in 0.5 ml of phosphate-buffered saline (PBS; pH 9.12) and sonicated for 40 s by means of an ultrasonic Cup Horn water bath. A total of 7.5 μl of FLUOS solution was added and mixed with bacteria at 4°C for 30 min. Fluorescein-stained M. bovis BCG (fluos-BCG) was pelleted (10,000 × g for 15 min), washed with 1.0 ml of PBS (pH 9.12), resuspended in Dulbecco's modified Eagle's medium (DMEM), and sonicated for 40 s. Fluos-BCG was prepared fresh for each experiment, and the number of CFU was determined by plating serial 10-fold dilutions on 7H10 agar plates. FLUOS labeling did not affect the viability of mycobacteria. Alveolar macrophages were infected overnight (37°C) with fluos-BCG (multiplicity of infection [MOI], 3:1 to 5:1) and subsequently treated with (or without) IFN-γ for 24, 48, and 72 h. After infection, cells were stained for surface MHC-II and B7.1 as described below and analyzed by flow cytometry. Where indicated, infected cells were gated on FL1 and examined for coexpression of MHC-II or B7.1. Ungated cells were also analyzed to determine the degree of phagocytosis.

Antibodies and cytokines.The following fluorescein isothiocyanate (FITC)-conjugated, phycoerythrin (PE)-conjugated, and biotinylated antibodies (identified by catalogue number) were purchased from PharMingen (San Diego, Calif.): biotin-mouse immunoglobulin G2a(κ) [IgG2a(κ)] (no. 553455), biotin-mouse antimouse I-Ab (no. 06042D), PE-hamster IgG2(λ) (no. 553935), PE-hamster antimouse CD80 (B7.1) (no. 553769), biotin-hamster IgG2(λ) (no. 550084), biotin-hamster antimouse CD80 (B7.1) (no. 09602D), rat-antimouse CD16/CD32 Fc-Block (Fcγ III/II Receptor) (no. 553141), FITC-rat IgG2a(κ) (no. 553929), FITC-rat antimouse CD86 (B7.2) (no. 09274D), FITC-hamster IgG1(λ) antitrinitrophenol (no. 553953), FITC-hamster antimouse CD11c (no. 09704A), FITC-rat antimouse CD40 (no. 09664D), PE-rat IgG2b(κ) (no. 553989), PE-rat antimouse CD11b (Mac1) (no. 01715B), FITC-rat IgG1(κ) (no. 553923), and FITC-rat antimouse Mac3 (no. 01715D). PE-rat antimouse F4/80 (no. MCA497PE) and PE-rat IgG2b (no. MCA1125PE) were purchased from Serotec Inc. (Raleigh, N.C.). Streptavidin-red 670 (no. 19543-024) was purchased from GibcoBRL (Grand Island, N.Y.). Murine IFN-γ was purchased from R & D Systems (Minneapolis, Minn.).

Isolation of murine alveolar macrophages.Age-matched mice were anesthetized with a lethal dose of tribromoethanol (240 mg/kg of body weight), and alveolar macrophages were isolated as described previously (13). The abdominal cavity was incised, mice were exsanguinated by transecting the left renal artery, and the chest was decompressed by a transverse incision at the level of the xiphoid. The trachea was exposed aseptically, and an 18-gauge Angiocath intravenous catheter (Becton Dickinson, Sandy, Utah) was inserted through a 1-mm incision at the first tracheal ring. A total of 3.6 ml of PBS (Sigma, St. Louis, Mo.) was instilled in nine separate 0.4-ml bronchoalveolar lavages (BAL). Alveolar macrophages were centrifuged at 2,000 × g for 10 min at 4°C and resuspended in DMEM (BioWhittaker, Walkersville, Md.) supplemented with 10% fetal calf serum, 100 IU of penicillin per ml, 100 μg of streptomycin per ml, 100 mM HEPES, 1× nonessential amino acids, 2.0 mM l-glutamine, and 0.05 mM 2-mercaptoethanol. Cell viability was routinely greater than 95% as determined by trypan blue (0.4%) exclusion. Cytospin slides of 2.5 × 104 cells/BAL/mouse were prepared by using a Cytospin 3 centrifuge (600 rpm for 6 min) (Shandon, Pittsburgh, Pa.) and stained with Diff-Quik (Dade Diagnostics, Aguada, Puerto Rico) for differential cell counts. Resident BAL cells were comprised of alveolar macrophages (90 to 95%), lymphocytes (5 to 10%), and occasional granulocytes (<1 to 2%).

Purification of the 19-kDa lipoprotein of M. tuberculosis (H37Ra).Stock cultures of M. tuberculosis were grown to log phase in Middlebrook 7H9 medium (Difco, Detroit, Mich.) supplemented with albumin, dextrose, and catalase (Difco). After 2 to 3 weeks, cultures were supplemented with glycerol (final volume, 6% [vol/vol]), thoroughly mixed, aliquoted, and frozen at −70°C. The M. tuberculosis H37Ra 19-kDa lipoprotein was isolated as described previously (32, 34). M. tuberculosis H37Ra was cultured to mid-log phase in 4 to 6 liters of 7H9 medium supplemented with albumin, dextrose, and catalase. Bacteria were pelleted in 500-ml tubes spun for 20 min at 8,000 rpm (5,220 × g) by a Sorvall RC-5B rotor. Bacterial pellets were combined and resuspended in 20 to 30 ml of lipopolysaccharide-free deionized water containing EDTA (7.5 mM), leupeptin (0.7 μg/ml), phenylmethylsulfonyl fluoride (0.2 mM), pepstatin A (0.7 μg/ml), DNase (10 U/ml), and RNase A (25 U/ml) (Boehringer-Mannheim, Indianapolis, Ind.). The solution was passed three times through a French press and centrifuged for 2 h at 100,000 × g and 4°C. Next, mycobacterial-cell-wall-containing pellets were resuspended in cold extraction buffer (pH 7.5) containing 20% Triton X-114 (Sigma) in Tris-HCl (50 mM) and NaCl (150 mM). After mixing overnight at 4°C, the cell wall preparation was centrifuged (100,000 × g) for 2 h at 4°C, and the remaining insoluble pellet was discarded. The supernatant was warmed to 37°C and centrifuged (2,400 × g) for 10 to 15 min at 37°C to separate aqueous and TX-114 layers. The upper aqueous layer was discarded, and the TX-114 layer was remixed and washed three to five times with an equal volume of cold buffer containing Tris-HCl (50 mM) and NaCl (150 mM). After washing, TX-114 soluble proteins were precipitated overnight (at −20°C) with 10 volumes of acetone (Fisher Chemicals, Fair Lawn, N.J.). Precipitated proteins were centrifuged (2,400 × g) for 20 to 30 min at 4°C, washed with cold acetone (−20°C), pelleted, and air dried at room temperature. The pellet was solubilized in reducing sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis sample buffer (62.5 mM Tris [pH 6.8], 2% SDS, 10% glycerol, 0.7 M 2-mercaptoethanol, and 0.01 μg of bromophenol blue per ml) and electroeluted through an SDS-12% polyacrylamide gel by using a Bio-Rad Model 491 Prep Cell as previously described (32). Fractions enriched for the M. tuberculosis H37Ra 19-kDa lipoprotein were identified by Western blotting, pooled, and reextracted with TX-114. After reprecipitation with acetone, the pellet was solubilized in 5 mM HEPES buffer (pH 7.0) containing 90% dimethyl sulfoxide, aliquoted, and stored at −70°C. Protein content was determined by using a detergent-compatible protein assay (Bio-Rad). Working 19-kDa lipoprotein preparations (50 μg/ml) were 85 to 90% pure and primarily contaminated with lipomannan and lipoarabinomannan (data not shown). Electroelution fractions collected before and after elution of the 19-kDa lipoprotein were used as negative control fractions and did not significantly affect IFN-γ-induced MHC-II expression or antigen processing and presentation (data not shown). Electroelution fractions contained less than 20 pg of lipopolysaccharide per ml as measured by Limulus amoebocyte lysate assay (E-Toxate kit; Sigma).

Staining and flow cytometry of alveolar macrophages.Alveolar macrophages (5 × 104) were resuspended in 0.015 ml of Fc-block (at a concentration of 2 μg/ml in 0.1% bovine serum albumin [BSA] in PBS) for 15 min on ice. Cocktails of primary antibodies or isotype control antibodies were diluted (for biotinylated MHC-II [I-Ab], 1:100; for B7.1, 1:50; for B7.2, 1:25; for CD40, 1:25; for CD11c, 1:25; for Mac3, 1:25; and for biotinylated F4/80, 1:25), and 0.030 ml was added to cells for 20 min on ice. Cells were washed with 5% BSA in PBS (pH 7.4), stained with 0.025 ml of streptavidin-red 670 (1:100 in 0.1% BSA) for 15 min on ice, washed with 5% BSA in PBS, and fixed with 1% paraformaldehyde in PBS (pH 7.4). Samples were stored in the dark at 4°C for subsequent analysis by flow cytometry.

Flow cytometry was performed with a Becton Dickinson FACScan flow cytometer (San Jose, Calif.). Forward light scatter, side light scatter, FL1 (FITC), FL2 (PE), and FL3 (streptavidin-red 670) settings were determined empirically to maximize the difference between isotype and specific staining. A total of 5,000 to 10,000 ungated events were collected. Histogram, dot plot, and mean fluorescence intensity (MFI) analyses were performed by using Cellquest (Becton Dickinson) software. Differences in mean specific MFI (MFI − isotype MFI ± standard error of the means [SEM]) values were statistically analyzed where indicated. In some cases (as indicated) where cells were not uniformly positive for surface staining, differences in mean specific geometric MFI (geoMFI) were analyzed.

Antigen processing and presentation assay.Alveolar macrophages were harvested as described above, plated (65,000 cells/well) into flat bottom 96-well plates (Falcon 35-3072) in complete medium and allowed to adhere overnight at 37°C. Alveolar macrophages were washed five times and pretreated with complete medium with or without the 19-kDa lipoprotein (250 ng/ml). After 24 h fresh medium containing murine IFN-γ (2 ng/ml) was added with or without fresh 19-kDa lipoprotein (250 ng/ml). After 48 h of incubation, alveolar macrophages were washed extensively with warm DMEM to remove residual IFN-γ and 19-kDa lipoprotein. Next, alveolar macrophages were incubated for 2 h with 300 to 5,000 μg of ovalbumin (OVA; Sigma) per ml, washed five times with warm DMEM, and allowed to process OVA overnight. After fixation with 1% paraformaldehyde (15 min) and extensive washing, OVA peptide (OVA323-339)-specific (18), MHC-II-restricted T-hybridoma cells (DOBW) were added (100,000/0.2 ml/well) and allowed to incubate overnight as described previously (31). Culture supernatants were removed after 18 to 24 h and stored at −70°C until they were assayed for interleukin 2 (IL-2). IL-2 was measured by using the standard colorimetric CTLL bioassay with Alamar Blue (Alamar Biosciences, Sacramento, Calif.) and a Bio-Rad 550 microplate reader as previously described (18). The CTLL assay was capable of detecting 20 to 100 pg of IL-2 per ml, which typically corresponds to 0.05 to 0.500 relative absorbance units (the optical density at 550 nm [OD550]). At each antigen concentration, the inhibition of antigen processing and presentation was determined by calculating the percent decrease in the OD550 between control cells and cells treated with the 19-kDa lipoprotein [percent inhibition = (control OD550 − experimental OD550)/control OD550 × 100].

Quantitative real-time reverse transcription-PCR for CIITA and MHC-II mRNA.Alveolar macrophages were isolated and first cultured overnight (18 to 24 h) without stimulation. After 24 h, cells were incubated with or without 19-kDa lipoprotein (500 ng/ml) for 24 h. Next, IFN-γ (2 ng/ml) with or without fresh 19-kDa lipoprotein (500 ng/ml) was added, and the cells were incubated for an additional 24 h. Afterwards, cells were pelleted (2,000 × g for 10 min), resuspended in lysis buffer, and passed through a QiaShredder according to the manufacturer's instructions (QIAGEN, Valencia, Calif.). Cellular RNA was purified by using the RNeasy kit (QIAGEN) according to the manufacturer's instructions. RNA (1 μg) was reverse transcribed by the SuperScript preamplification system (Life Technologies) as previously described (34). The cDNA mixture was diluted 1:5 and 10 μl was used for each gene-specific amplification reaction using a high-speed thermal cycler (I-Cycler; Bio-Rad, Hercules, Calif.). PCR products were detected by iQ-SYBR Green Supermix (Bio-Rad). The amplification cycle was 95°C for 10 s, 52°C for 15 s, and 72°C for 20 s (35 cycles). Primers were designed by using OLIGO version 6.4 (Molecular Biology Insights, Cascade, Colo.) and are shown as follows: for GAPDH (glyceraldehyde-3-phosphate dehydrogenase), 5′-AACGACCCCTTCATTGAC-3′ (sense) and 5′-TCCACGACATACTCAGCAC-3′(antisense) (predicted size, 191 bp); for total class II transactivator (CIITA), 5′-ACGCTTTCTGGCTGGATTAGT-3′ (sense) and 5′-TCAACGCCAGTCTGACGAAGG-3′ (antisense) (predicted size, 342 bp); and for MHC-II (I-Ab α-chain), 5′-GCTCCTCAAGCGACTGTG-3′ (sense) and 5′-AAGCTGGTCTCATAAACACCG-3′ (antisense) (predicted size, 155 bp). cDNA copy numbers for each gene were quantified by using standard curves of known quantities of gel-purified PCR products prepared with a QiaQuick gel extraction kit. Standard curves of the PCR threshold cycle number versus cDNA copy number were generated and used to quantify the corresponding CIITA and MHC-II mRNA copy numbers in alveolar macrophages treated as described. The copy numbers of CIITA and MHC-II in both control and treated alveolar macrophages were normalized to GAPDH expression.

Statistical analysis.The Wilcoxon rank sum test or the Student's t test was used where indicated. Significant differences are indicated (P ≤ 0.05).

RESULTS

Murine alveolar macrophages constitutively express surface MHC-II, CD80 (B7.1), and CD11c.Alveolar macrophages comprise more than 90 to 95% of alveolar cells and are the first immune effector cells to encounter aerosolized mycobacteria transmitted to alveolar spaces. By using flow cytometry, resident alveolar macrophages from healthy normal mice were analyzed for the expression of MHC-II and the costimulatory molecules CD80 (B7.1) and CD86 (B7.2) essential for the activation of CD4+ T cells. In Fig. 1A to F, representative histograms show the degree of surface MHC-II, B7.1, B7.2, CD11b (Mac-1), CD11c, and CD40 expression on alveolar macrophages isolated and pooled from age-matched mice. Mean specific MFIs (± SEM) from five different experiments also are shown on the figure. Resident alveolar macrophages expressed B7.1 (panel A), MHC-II (panel D), CD11c (panel E), and FcRIII/II (data not shown) constitutively but did not express B7.2, CD11b, and CD40 (panels B, C, and F, respectively). In addition, expression of Mac-3 or F4/80 was not detected (data not shown). However, CD11b was expressed on alveolar macrophages cultured overnight in vitro (data not shown). Thus, resident alveolar macrophages were characterized by the expression of MHC-II, B7.1, CD11c, and FcRIII/II. By means of two-color analysis, resident alveolar macrophages were also analyzed for coexpression of B7.1 and MHC-II (panel H) and CD11c (panel J) molecules. We determined that the majority (85.2% ± 1.3%) of resident alveolar macrophages coexpressed B7.1 and CD11c. In addition, 20.3% ± 3.2% also coexpressed MHC-II (panel H), thereby characterizing the two major subsets of resident alveolar macrophages as MHC-II+/CD11c+/B7.1+ and MHC-II−/CD11c+/B7.1+. In addition, a small proportion of bronchoalveolar cells expressed MHC-II only (7.6% ± 1.0%), and 7.26% ± 0.45% of cells did not stain for either surface B7.1 or MHC-II. Thus, a subset of resident alveolar macrophages expresses the molecules necessary for CD4+-T-cell recognition and activation.

FIG. 1.
  • Open in new tab
  • Download powerpoint
FIG. 1.

Resident alveolar macrophages express CD80 (B7.1), CD11c, and MHC-II. (A to F) Alveolar macrophages were isolated from 8- to 10-week-old female C57BL/6 mice and stained as described in Materials and Methods. A total of 5,000 events per sample was analyzed by flow cytometry. Histograms show both specific staining (shaded) and isotype control staining (unshaded) for CD80 (B7.1), CD86 (B7.2), CD11b (Mac-1), CD11c, CD40, and MHC-II. Data shown on the panels are the mean specific MFIs (± SEM) from five independent experiments demonstrating the expression of B7.1, MHC-II, and CD11c (for FcRIII/II, MFI = 16.9 ± 7.1; data not shown). B7.2, Mac-1, and CD40 were not detected. (G to J) Dot blots show coexpression of MHC-II and CD11c on B7.1-expressing alveolar macrophages. The mean percentages (± SEM) of total cells expressing B7.1 and coexpressing MHC-II and CD11c from five independent experiments are shown on the panels inserts. Staining with isotype control antibodies was less than 2 to 3% of the total events detected in any single quadrant.

IFN-γ enhances surface MHC-II molecule expression on alveolar macrophages.IFN-γ regulates the microbicidal and antigen-presenting functions of macrophages and, thus, has a critical role in protective immunity to mycobacteria (11, 12). Alveolar macrophages were cultured either with or without IFN-γ and analyzed for the expression of MHC-II and B7.1. In Fig. 2, representative histograms demonstrate IFN-γ-enhanced MHC-II expression (panels A to C). Expression of B7.1 (panels D to F), B7.2 (data not shown), and CD11b (data not shown) was not effected by IFN-γ. Since the staining of untreated (control) cells was not uniform, mean specific geoMFI was used to quantify the increased expression of MHC-II. In panel G, the values of the mean specific geoMFI (± SEM) for MHC-II expression from six independent experiments are summarized. Equivalent levels of MHC-II were detected on freshly isolated alveolar macrophages (15.3 ± 1.2) (Fig. 1D) and alveolar macrophages cultured for 24 (17.8 ± 2.0), 48 (16.7 ± 2.5), and 72 h (14.9 ± 1.5) in medium alone (Fig. 2A to C). After 24 h, IFN-γ treatment significantly enhanced MHC-II expression by 1.5-fold (17.8 ± 4.9 versus 26.7 ± 6.9; P < 0.05). In addition, after 48 or 72 h of culture, IFN-γ treatment resulted in a significant increase in MHC-II expression levels of threefold (16.7 ± 2.5 versus 46.4 ± 5.5) and sixfold (14.9 ± 1.4 versus 83.7 ± 10.2), respectively, compared to controls (n = 6, P ≤ 0.05).

FIG. 2.
  • Open in new tab
  • Download powerpoint
FIG. 2.

IFN-γ enhances expression of MHC-II on alveolar macrophages. (A to F) Alveolar macrophages were isolated from mice and cultured at the indicated times in the presence (shaded) or absence (dark black line; unshaded) of murine IFN-γ (2 ng/ml). Cells were stained for MHC-II and B7.1 as described, and histograms from a representative experiment (n = 5) are shown. Specific staining in the absence and presence of IFN-γ are shown and compared to isotype control staining. In panel G, the mean specific geoMFI (± SEM) for MHC-II from five experiments is shown. The Wilcoxon rank sum test was used to compare MHC-II expression between control and IFN-γ-stimulated alveolar macrophages. Statistically significant differences in specific geoMFI are designated by an asterisk (P ≤ 0.05).

Two-color analysis determined the extent of MHC-II coexpression on B7.1+ and B7.1− alveolar cells. In Fig. 3, representative dot blots for alveolar macrophages cultured with (panels A to C) or without (panels D to F) IFN-γ show the effect of IFN-γ on MHC-II expression, with data summarizing six independent experiments (mean percentage ± SEM). In the absence of IFN-γ, the proportion of B7.1- and MHC-II-coexpressing cells increased somewhat after 24 h (35.2% ± 1.7 in panel A versus 20.3% ± 3.2 in Fig. 1, panel H) but did not change further after 48 to 72 h (range, 27.9 to 35.2%; panels A to C). This result suggested that overnight culture conditions increased the proportion of cells expressing MHC-II by 10 to 15%, which was not discernible by single-color analysis (Fig. 1D and 2G). In the presence of IFN-γ (Fig. 3, panels D to F), the proportion of B7.1+/MHC-II+ cells increased markedly at each time point studied (P < 0.05 compared to untreated cells in panels A to C). After 72 h of IFN-γ treatment, the proportion of B7.1+/MHC-II+ cells increased from 52.7% ± 3.8% (panel D) to 67.8% ± 4.6% (panel F), corresponding to a concomitant decrease in the proportion of B7.1+/MHC-II− cells (32.2% ± 4.1%, as seen in panel D, versus 7.7% ±1.7%, as seen in panel F; P < 0.05). After 72 h, nearly 90% of all B7.1+ cells coexpressed MHC-II. Although enhanced MHC-II expression occurred primarily on B7.1+ alveolar macrophages, 10 to 15% of total MHC-II was also detected on B7.1-negative cells. Thus, localized expression of IFN-γ in alveolar spaces may enhance MHC-II expression and the ability of alveolar macrophages to present antigen and activate CD4+ T cells.

FIG. 3.
  • Open in new tab
  • Download powerpoint
FIG. 3.

Enhanced coexpression of MHC-II on IFN-γ-treated B7.1+ alveolar macrophages. (A to F) Alveolar macrophages were isolated, cultured at the indicated times with IFN-γ (2 ng/ml) or without IFN-γ and then stained for B7.1 and MHC-II. A total of 5,000 to 7,500 events were collected for two-color analysis and representative dot blots are shown. Quadrant settings were used to calculate the distribution of cell types within the entire (ungated) alveolar macrophages population. The levels of expression of MHC-II and B7.1 from six different experiments are summarized and shown on panels (mean % total ± SEM) for upper- and lower-right quadrants. Isotype control staining was less than 2 to 3% in each quadrant.

M. bovis infection and the 19-kDa lipoprotein inhibit IFN-γ-mediated upregulation of MHC-II on alveolar macrophages.Since alveolar macrophages are phagocytic for M. bovis BCG and IFN-γ enhances expression of MHC-II on alveolar macrophages, we next studied IFN-γ-inducible MHC-II expression in alveolar macrophages infected with BCG. First, phagocytosis of M. bovis BCG by alveolar macrophages was analyzed by infecting alveolar macrophages overnight at a low MOI (3 to 5) with fluos-BCG. Figure 4 (panel A) demonstrates that at 24 h after infection, the change in MFI between uninfected and fluos-BCG-infected alveolar macrophages was 80.8 ± 7.2 and indicated that 47.8% ± 7.2% of alveolar macrophages had phagocytosed fluos-BCG (n = 6). Additional analysis of MHC-II expression on fluos-BCG-infected B7.1+ cells (R1 gated, panel B) indicated that there was a similar uptake of fluos-BCG by both MHC-II−/B7.1+ and MHC-II+/B7.1+ alveolar macrophages (45.8% ± 3.5% and 49.7% ± 3.1%, respectively). Uptake and viability of BCG were unaffected by fluorescein labeling (data not shown).

FIG. 4.
  • Open in new tab
  • Download powerpoint
FIG. 4.

Phagocytosis of M. bovis BCG by alveolar macrophages. Alveolar macrophages were incubated overnight with fluos-BCG at a low MOI (3 to 5) and analyzed by flow cytometry. (A) A representative histogram shows phagocytosis of fluos-BCG. (B) MHC-II and B7.1 expression on fluos-BCG-positive cells (region R1, panel A) from a representative experiment reveals similar uptake of bacteria by both MHC-II+ and MHC-II− B7.1+ alveolar macrophages.

Second, to determine whether IFN-γ-induced MHC-II expression was affected by M. bovis BCG infection, cells were infected overnight at a low MOI and then stimulated with IFN-γ for 24, 48 or 72 h. As shown in a representative experiment (Fig. 5), IFN-γ treatment (panel A) enhanced MHC-II expression (two- to threefold) in both infected and uninfected control cells. However, after 72 h (panel B), M. bovis BCG infection inhibited upregulation of MHC-II by IFN-γ. In four independent experiments, a 47.8% ± 11.9% reduction in MHC-II was measured (P < 0.05, by paired Student's t test). In contrast, infection of alveolar macrophages in the absence of IFN-γ did not affect surface MHC-II expressed constitutively on the 20 to 30% of resident alveolar macrophages described in Fig. 1 (panel H) and Fig. 3 (panels A to C) (data not shown).

FIG. 5.
  • Open in new tab
  • Download powerpoint
FIG. 5.

M. bovis BCG infection and a 19-kDa mycobacterial lipoprotein inhibit IFN-γ-inducible MHC-II expression on alveolar macrophages. (A to D) Alveolar macrophages were cultured with or without BCG at an MOI of 3 to 5 and with or without the 19-kDa lipoprotein (250 ng/ml) for 24 h. IFN-γ (2 ng/ml) was added after infection or pretreatment with the 19-kDa lipoprotein, and alveolar macrophages were cultured for an additional 24, 48, and 72 h in the presence of IFN-γ. The expression of MHC-II was measured by flow cytometry, and histograms representative of four independent experiments are shown. IFN-γ-induced MHC-II expression on BCG-infected or 19-kDa-lipoprotein-treated macrophages (black lines) is compared to control treated cells (shaded) after 48 and 72 h of IFN-γ exposure. Isotype control staining for both treatment groups is shown with overlapping thin lines. (E to F) Alveolar macrophages were isolated, cultured overnight, and then treated without (medium) or with the 19-kDa lipoprotein (500 ng/ml) for 24 h. After 24 h, fresh 19-kDa lipoprotein was added along with IFN-γ (2 ng/ml), and cells were incubated for an additional 48 h. CIITA and MHC-II mRNA expression was measured by real-time PCR and normalized to GAPDH expression. Data from a representative experiment (n = 3) are shown. Decreases in CIITA and MHC-II mRNA expression were analyzed by using a paired Student's t test, and significant differences are designated with asterisks (P ≤ 0.05).

Recent studies have identified the 19-kDa lipoprotein of M. bovis BCG and M. tuberculosis as a TLR-2 ligand that modulates APC function (2, 32). As shown in Fig. 5C and D, the 19-kDa lipoprotein decreased the MFI for MHC-II by 69.8 ± 10.4 units (panel D) compared to the MFI of control cells at 72 h (P ≤ 0.05 by paired Student's t test). In five independent experiments, after 72 h a 35.5% ± 4.6% reduction in MHC-II was observed compared to MHC-II in cells at 24 h (P ≤ 0.05 by Wilcoxon rank sum). Thus, both whole bacilli and the 19-kDa lipoprotein, a major cell wall component of M. bovis BCG and M. tuberculosis, inhibited IFN-γ-inducible MHC-II expression on alveolar macrophages.

To determine the potential mechanisms for the 19-kDa lipoprotein-mediated inhibition of surface MHC-II expression, we also analyzed transcription of CIITA mRNA and MHC-II mRNA. RNA was isolated from alveolar macrophages pretreated with the 19-kDa lipoprotein (500 ng/ml) 24 h before IFN-γ treatment. After 24 h of IFN-γ treatment, mRNA for CIITA and MHC-II (I-Ab, beta chain) was measured by real-time reverse transcription-PCR. As shown in a representative experiment (Fig. 5E and F), the 19-kDa lipoprotein significantly decreased (P ≤ 0.05) the copy numbers of CIITA (panel E) and MHC-II (panel F) mRNA by 35.1 and 68.3%, respectively. In three independent experiments, a 41.9% ± 8.5% (mean ± SEM) reduction in CIITA mRNA and a 51.5% ± 19.7% (mean ± SEM) reduction in MHC-II mRNA were detected. Depletion of MHC-II mRNA correlated with the delayed loss of surface MHC-II detected after 72 h (Fig. 5B and D) when sufficient decay of surface MHC-II had occurred. Thus, the 19-kDa lipoprotein inhibited IFN-γ-induced transcription of both CIITA mRNA and MHC-II mRNA, resulting in decreased surface MHC-II.

Inhibition of MHC-II expression and antigen processing and presentation by the 19-kDa lipoprotein are dependent on TLR-2.In preliminary experiments we had determined that MHC-II on alveolar macrophages was capable of binding and presenting peptide to MHC-II-restricted DOBW (data not shown). To determine if changes in MHC-II expression (Fig. 5D) affected antigen processing, the effect of the 19-kDa lipoprotein on processing and presentation of ovalbumin was tested with an MHC-II-restricted T-cell DOBW. In Fig. 6, a representative experiment demonstrates that recognition of MHC-II-peptide complexes in normal alveolar macrophages pretreated with the 19-kDa lipoprotein and pulsed with ovalbumin at concentrations of 316, 1,000, or 3,160 μg/ml was significantly (P ≤ 0.05) inhibited by 77.2, 47.6, and 23.2%, respectively. In three different experiments, the average (± SEM) degree of inhibition was 85.8% ± 7.2%, 57.5% ± 8.7%, and 29.6% ± 7.0%.

FIG. 6.
  • Open in new tab
  • Download powerpoint
FIG. 6.

The 19-kDa mycobacterial lipoprotein inhibits antigen processing and presentation in alveolar macrophages. Alveolar macrophages were harvested, plated in duplicate in 96-well flat bottom plates (65,000 cells/well), and incubated overnight. Adherent cells were washed and incubated for 24 h with or without 19-kDa lipoprotein. Next, cells received IFN-γ (2 ng/ml) in fresh medium (controls) or medium supplemented with fresh 19-kDa lipoprotein. After 48 h, cells were washed extensively and pulsed for 2 h with ovalbumin, and antigen presentation was measured 24 h later as described. Ovalbumin-specific DOBW responses were measured, and mean (± standard deviation) DOBW responses from a representative experiment (n = 3) are shown. Percent inhibition was calculated as the reduction in the OD550 measured by the CTLL assay for IL-2 (see Materials and Methods). Statistically significant inhibition (P ≤ 0.05) of ovalbumin processing was measured at each antigen dose by using a paired Student's t test.

Next we determined whether TLR-2 was necessary for the 19-kDa lipoprotein to inhibit MHC-II expression and antigen processing. Thus, we analyzed IFN-γ-inducible expression of MHC-II in the presence and absence of the 19-kDa lipoprotein in alveolar macrophages isolated from TLR-2 −/− and wild-type mice. In Fig. 7, representative histograms of three independent experiments show that the specific MFI for MHC-II expression was reduced 41.6% ± 4.9% (P ≤ 0.05) in wild-type alveolar macrophages after 72 h (panel A). In contrast, expression of IFN-γ-inducible MHC-II in alveolar macrophages isolated from TLR-2−/− mice (panel B) was unaffected by the 19-kDa lipoprotein, suggesting that intact TLR-2 was necessary for the inhibition of MHC-II expression mediated by the 19-kDa lipoprotein. In addition, as shown in Fig. 7C, the 19-kDa lipoprotein did not inhibit processing of ovalbumin by alveolar macrophages obtained from TLR-2−/− mice. Although it is not known why TLR-2 −/− alveolar macrophages appeared to process and present ovalbumin more efficiently than wild-type cells, the 19-kDa lipoprotein-mediated inhibition of MHC-II expression and antigen processing by wild-type alveolar macrophages was dependent upon TLR-2.

FIG. 7.
  • Open in new tab
  • Download powerpoint
FIG. 7.

Inhibition of MHC-II expression and antigen processing by the 19-kDa lipoprotein is dependent on TLR-2. (A to B) Alveolar macrophages were isolated from age-matched C57BL/6 wild-type and C57BL/6 TLR-2−/− mice. After overnight incubation with (dark lines) or without (shaded) the 19-kDa lipoprotein (250 ng/ml), alveolar macrophages were treated for 48 h with IFN-γ (2 ng/ml) and then analyzed for surface MHC-II expression. Histograms representative of three different experiments are shown. (C) Alveolar macrophages were isolated from wild-type C57BL/6 and TLR-2 −/− mice, pretreated with the 19-kDa lipoprotein or medium, and then incubated for 48 h with IFN-γ with or without fresh 19-kDa lipoprotein or medium. Next, cells were pulsed with ovalbumin, and DOBW responses were analyzed as described in the legend of Fig. 6. Mean (± standard deviation) DOBW responses for wild-type alveolar macrophages and TLR-2 −/− alveolar macrophages pretreated with medium or 19-kDa lipoprotein from a representative experiment (n = 3) are shown. By a paired Student's t test, statistically significant inhibition (P ≤ 0.05) was detected at each ovalbumin dose in wild-type cells pretreated with the 19-kDa lipoprotein. Antigen processing and presentation were not inhibited in TLR-2 −/− cells pretreated with the 19-kDa lipoprotein.

DISCUSSION

Alveolar macrophages are an important line of defense against aerosolized microorganisms and are bactericidal for many phagocytosed organisms, but their role in activating T cells toward intracellular pathogens remains undefined (7, 53). The present study was undertaken to determine the phenotype and function of murine alveolar macrophages as APC. Resident alveolar macrophages constitutively expressed the molecules necessary for antigen processing and activation of CD4+ T cells. The majority (85%) of alveolar macrophages expressed B7.1 (CD80) and CD11c but not B7.2 (CD86), CD11b (Mac-1), CD40, and Mac-3. In addition, a substantial proportion of alveolar macrophages expressed MHC-II. Recently, other investigators have also noted B7.1, CD11c, and MHC-II expression on alveolar macrophages (35, 43). Although we did not detect F4/80, a low level of F4/80 expression on alveolar macrophages has been reported by others and may differ between different mouse strains (24, 29). In addition, CD11b was not found on freshly isolated cells but was rapidly upregulated on alveolar macrophages cultured for 24 h and unaffected by IFN-γ (data not shown) (44). Coexpression of CD11b, CD11c, CD80, and MHC-II suggested that cultured alveolar macrophages might have a dendritic cell phenotype; however, their morphology was not typical of dendritic cells, and they did not express CD8 (15, 22). A recent study with carboxyfluorescein diacetate succinimidyl ester-labeling of lung cells suggested that, based on CD11c expression, 14% of resident lung cells might be dendritic cells (26). However, alveolar macrophages that express CD11c were not distinguished from parenchymal macrophages, and thus the number of lung dendritic cells may have been overestimated.

IFN-γ, a critical cytokine for immunity against intracellular pathogens, enhances both the microbicidal and antigen-presenting functions of macrophages (6, 11, 41). Also, IFN-γ is expressed in alveolar spaces and lung parenchyma as early as 10 days after mycobacterial infection (13, 14). In the present study, IFN-γ markedly increased expression of MHC-II on alveolar macrophages, such that after 72 h the majority of B7.1+ alveolar macrophages coexpressed MHC-II. IFN-γ did not alter the expression of B7.1. IFN-γ also enhanced the processing and presentation of soluble protein antigen to MHC-II-restricted T cells, indicating that alveolar macrophages are competent APC. This finding contrasts with other studies of human and murine alveolar macrophages that suggested that alveolar macrophages inhibit T-cell activation (40, 48). Generally, these studies measured the effects of alveolar macrophages on mitogen-driven CD4+-T-cell responses in non-MHC-matched experimental systems and did not directly address antigen presentation to MHC-II-restricted T cells. Our studies did not address the ability of alveolar macrophages to activate naïve CD4+ T cells but do indicate that these macrophages, particularly after IFN-γ activation, can process antigen for memory CD4+-T-cell activation.

Murine models have demonstrated that the lung is uniquely susceptible to mycobacterial infection. Compared to intravenous or intraperitoneal challenge, many fewer aerosolized organisms are needed to establish progressive infection in murine lungs (9). When aerosolized M. tuberculosis or M. bovis BCG cells are deposited in alveolar spaces and phagocytosed by alveolar macrophages, they continue to grow in the lung until adaptive immunity controls the infection (5, 13). Mycobacteria reside within alveolar and parenchymal macrophages in the lung but antigen-specific CD4+-T-cell responses fail to eradicate all bacteria. Mechanisms used by M. tuberculosis for its survival in macrophages are starting to be defined. Recent studies from our laboratory have shown that murine bone marrow-derived macrophages infected with M. tuberculosis or exposed to the 19-kDa lipoprotein were inhibited in MHC-II antigen processing and presentation (31). The inhibition of MHC-II function was dependent on TLR-2 and required prolonged exposure to the 19-kDa-lipoprotein (32). In the present study, we found that alveolar macrophages infected with M. bovis BCG or exposed to the 19-kDa-lipoprotein were also inhibited in IFN-γ-mediated upregulation of MHC-II expression. However, the degree of inhibition was not as marked and may reflect the terminally differentiated status of alveolar macrophages. Nonetheless, the inhibition was associated with a significant decrease in both CIITA mRNA and MHC-II mRNA expression as reported for other experimental systems (23, 34, 47). Although mycobacterial growth is not controlled in IFN-γ gene knockout mice, antigen-specific T-cell activation and delayed-type hypersensitivity responses can be detected, indicating that IFN-γ-independent mechanisms may evoke MHC-II-restricted responses (37). The inhibition of MHC-II expression by mycobacterial lipoproteins is only one component of host immune responses modulated during mycobacterial infection. Furthermore, it is not known how mycobacterial lipoproteins affect the functions of peripheral monocytes newly recruited to sites of mycobacterial infection where extracellular lipoproteins may be present (1). Additional studies that examine in vivo expression of MHC-II on infected and uninfected macrophages should help determine more directly how mycobacterial lipoproteins influence antigen presentation, T-cell activation, and mycobacterial persistence in the lung. Nonetheless, we demonstrated that IFN-γ-dependent processing and presentation of protein antigen to MHC-II-restricted T cells by alveolar macrophages were inhibited through a TLR-2-dependent mechanism. Other studies from our laboratory have shown that the inhibition of IFN-γ signaling by the 19-kDa lipoprotein in bone marrow-derived macrophages does not involve STAT1 phosphorylation, is independent of the suppressor of cytokine signaling genes, but is associated with partial inhibition of USF-1 (23, 34).

The role of alveolar macrophages in host defenses against M. tuberculosis remains to be defined. Alveolar macrophage depletion in vivo can reduce mycobacterial growth, augment early T-cell activation, and increase the survival of mice (25). The effect of this depletion suggests that either the alveolar macrophage is ineffective in controlling mycobacteria and activating T cells or that the alveolar milieu limits the activation of macrophages. In vitro studies indicate that alveolar macrophages are mycobactericidal, and our results suggest that alveolar macrophages are competent APC for CD4+ T cells. Evidence for an inhibitory alveolar milieu is suggested by studies demonstrating that IL-10 or transforming growth factor β inhibits macrophage function (20, 42, 49). Furthermore, surfactant proteins A and D modulate the phagocytosis of M. tuberculosis, inhibit T-cell proliferation, and inhibit expression of tumor necrosis factor α, IL-10, and nitric oxide in human and rat macrophages (7, 8, 10, 36). Although mice deficient in surfactant proteins A and D are more susceptible to pyogenic bacterial infections, it is not yet known if these mice have impaired responses to mycobacterial infection (27, 28). Thus, susceptibility of the lung to mycobacterial infection involves both normal down-regulatory homeostatic functions in the lung and pathogen-driven inhibition of acquired immunity (e.g., inhibition of MHC-II).

The role of TLRs in mycobacterial immunity is the subject of intense research (46). The recognition of mycobacteria and mycobacterial antigens by TLRs induces proinflammatory cytokines (e.g., tumor necrosis factor α and IL-12) and nitric oxide synthase (30, 51). Although M. tuberculosis may contain pattern-associated molecular patterns for many different TLRs, its lipoproteins represent a predominant source of TLR-2 ligands (2). In our studies, prolonged stimulation of macrophages (>16 h) through TLR-2 inhibited IFN-γ-inducible MHC-II expression and antigen processing and presentation. These data contrast with other reports, where synthetic 19-kDa lipoproteins were shown to induce expression of MHC-II on immature human dendritic cells which may respond differently than terminally differentiated alveolar macrophages (19). However, other studies have reported that M. tuberculosis inhibited human dendritic cell maturation (17). We hypothesize that the mechanism of immune evasion requires that mycobacteria survive the initial activation of macrophages by mycobacterial pattern-associated molecular patterns so that persistent TLR-2 signaling can occur and interfere with IFN-γ-regulated MHC-II antigen processing. Indeed, recent data suggest that during chronic M. tuberculosis infection, the levels of MHC-II on lung macrophages were similar to levels detected in uninfected mice (16). Thus, TLR-2-dependent inhibition of IFN-γ effects and the inhibitory milieu of alveolar spaces limit inflammation and adaptive immunity and thus provide a mechanism for mycobacteria to persist in macrophages.

ACKNOWLEDGMENTS

We thank Osamu Takeuchi and Shizua Akira for permission to use the TLR-2 gene knockout mice provided by Douglas Golenbock (University of Massachusetts, Worcester, Mass.).

This work was supported by National Institutes of Health Grants K8-HL04299 (to S.A.F.), AI-35726, AI-34343, and AI-44794 (to C.V.H.), and HL55967, AI-27243, and AI-95383 (to W.H.B.). Antibodies used for flow cytometry were purchased through the Immune Function Core Facility of the Center for AIDS Research (NIH AI-36219) at Case Western Reserve University.

FOOTNOTES

    • Received 1 October 2003.
    • Returned for modification 12 November 2003.
    • Accepted 8 January 2004.
  • Copyright © 2004 American Society for Microbiology

REFERENCES

  1. 1.↵
    Beatty, W. L., E. R. Rhoades, H. J. Ullrich, D. Chatterjee, J. E. Heuser, and D. G. Russell. 2000. Trafficking and release of mycobacterial lipids from infected macrophages. Traffic1:235-247.
    OpenUrlCrossRefPubMedWeb of Science
  2. 2.↵
    Brightbill, H. D., D. H. Libraty, S. R. Krutzik, R. B. Yang, J. T. Belisle, J. R. Bleharski, M. Maitland, M. V. Norgard, S. E. Plevy, S. T. Smale, P. J. Brennan, B. R. Bloom, P. J. Godowski, and R. L. Modlin. 1999. Host defense mechanisms triggered by microbial lipoproteins through toll-like receptors. Science285:732-736.
    OpenUrlAbstract/FREE Full Text
  3. 3.↵
    Chensue, S. W., K. Warmington, J. Ruth, P. Lincoln, M. C. Kuo, and S. L. Kunkel. 1994. Cytokine responses during mycobacterial and schistosomal antigen-induced pulmonary granuloma formation. Production of Th1 and Th2 cytokines and relative contribution of tumor necrosis factor. Am. J. Pathol.145:1105-1113.
    OpenUrlPubMedWeb of Science
  4. 4.↵
    Constant, S. L., K. S. Lee, and K. Bottomly. 2000. Site of antigen delivery can influence T cell priming: pulmonary environment promotes preferential Th2-type differentiation. Eur. J. Immunol.30:840-847.
    OpenUrlCrossRefPubMedWeb of Science
  5. 5.↵
    Cooper, A. M., J. E. Callahan, M. Keen, J. T. Belisle, and I. M. Orme. 1997. Expression of memory immunity in the lung following re-exposure to Mycobacterium tuberculosis.Tuber. Lung Dis.78:67-73.
    OpenUrlCrossRefPubMedWeb of Science
  6. 6.↵
    Cooper, A. M., D. K. Dalton, T. A. Stewart, J. P. Griffin, D. G. Russell, and I. M. Orme. 1993. Disseminated tuberculosis in interferon-γ gene-disrupted mice. J. Exp. Med.178:2243-2247.
    OpenUrlAbstract/FREE Full Text
  7. 7.↵
    Crouch, E., K. Hartshorn, and I. Ofek. 2000. Collectins and pulmonary innate immunity. Immunol. Rev.173:52-65.
    OpenUrlCrossRefPubMedWeb of Science
  8. 8.↵
    Crouch, E. C. 2000. Surfactant protein-D and pulmonary host defense. Respir. Res.1:93-108.
    OpenUrlCrossRefPubMed
  9. 9.↵
    Dunn, P. L., and R. J. North. 1995. Virulence ranking of some Mycobacterium tuberculosis and Mycobacterium bovis strains according to their ability to multiply in the lungs, induce lung pathology, and cause mortality in mice. Infect. Immun.63:3428-3437.
    OpenUrlAbstract/FREE Full Text
  10. 10.↵
    Ferguson, J. S., and L. S. Schlesinger. 2000. Pulmonary surfactant in innate immunity and the pathogenesis of tuberculosis. Tuber. Lung Dis.80:173-184.
    OpenUrlCrossRefPubMed
  11. 11.↵
    Flynn, J. L., and J. Chan. 2001. Immunology of tuberculosis. Annu. Rev. Immunol.19:93-129.
    OpenUrlCrossRefPubMedWeb of Science
  12. 12.↵
    Flynn, J. L., J. Chan, K. J. Triebold, D. K. Dalton, T. A. Stewart, and B. R. Bloom. 1993. An essential role for interferon γ in resistance to Mycobacterium tuberculosis infection. J. Exp. Med.178:2249-2254.
    OpenUrlAbstract/FREE Full Text
  13. 13.↵
    Fulton, S. A., T. D. Martin, R. W. Redline, and W. Henry Boom. 2000. Pulmonary immune responses during primary Mycobacterium bovis- Calmette-Guerin bacillus infection in C57Bl/6 mice. Am. J. Respir. Cell Mol. Biol.22:333-343.
    OpenUrlCrossRefPubMedWeb of Science
  14. 14.↵
    Fulton, S. A., S. M. Reba, T. D. Martin, and W. H. Boom. 2002. Neutrophil-mediated mycobacteriocidal immunity in the lung during Mycobacterium bovis BCG infection in C57BL/6 mice. Infect. Immun.70:5322-5327.
    OpenUrlAbstract/FREE Full Text
  15. 15.↵
    Gonzalez-Juarrero, M., and I. M. Orme. 2001. Characterization of murine lung dendritic cells infected with Mycobacterium tuberculosis.Infect. Immun.69:1127-1133.
    OpenUrlAbstract/FREE Full Text
  16. 16.↵
    Gonzalez-Juarrero, M., T. S. Shim, A. Kipnis, A. P. Junqueira-Kipnis, and I. M. Orme. 2003. Dynamics of macrophage cell populations during murine pulmonary tuberculosis. J. Immunol.171:3128-3135.
    OpenUrlAbstract/FREE Full Text
  17. 17.↵
    Hanekom, W. A., M. Mendillo, C. Manca, P. A. Haslett, M. R. Siddiqui, C. Barry III, and G. Kaplan. 2003. Mycobacterium tuberculosis inhibits maturation of human monocyte-derived dendritic cells in vitro. J. Infect. Dis.188:257-266.
    OpenUrlCrossRefPubMed
  18. 18.↵
    Harding, C. V., D. S. Collins, O. Kanagawa, and E. R. Unanue. 1991. Liposome-encapsulated antigens engender lysosomal processing for class II MHC presentation and cytosolic processing for class I presentation. J. Immunol.147:2860-2863.
    OpenUrlAbstract
  19. 19.↵
    Hertz, C. J., S. M. Kiertscher, P. J. Godowski, D. A. Bouis, M. V. Norgard, M. D. Roth, and R. L. Modlin. 2001. Microbial lipopeptides stimulate dendritic cell maturation via Toll-like receptor 2. J. Immunol.166:2444-2450.
    OpenUrlAbstract/FREE Full Text
  20. 20.↵
    Hirsch, C. S., T. Yoneda, L. Averill, J. J. Ellner, and Z. Toossi. 1994. Enhancement of intracellular growth of Mycobacterium tuberculosis in human monocytes by transforming growth factor-β1. J. Infect. Dis.170:1229-1237.
    OpenUrlCrossRefPubMedWeb of Science
  21. 21.↵
    Kamijo, R., J. Le, D. Shapiro, E. A. Havell, S. Huang, M. Aguet, M. Bosland, and J. Vilcek. 1993. Mice that lack the interferon-gamma receptor have profoundly altered responses to infection with Bacillus Calmette-Guerin and subsequent challenge with lipopolysaccharide. J. Exp. Med.178:1435-1440.
    OpenUrlAbstract/FREE Full Text
  22. 22.↵
    Kelsall, B. L., C. A. Biron, O. Sharma, and P. M. Kaye. 2002. Dendritic cells at the host-pathogen interface. Nat. Immunol.3:699-702.
    OpenUrlCrossRefPubMedWeb of Science
  23. 23.↵
    Kincaid, E. Z., and J. D. Ernst. 2003. Mycobacterium tuberculosis exerts gene-selective inhibition of transcriptional responses to IFN-γ without inhibiting STAT1 function. J. Immunol.171:2042-2049.
    OpenUrlAbstract/FREE Full Text
  24. 24.↵
    Lagranderie, M., M. A. Nahori, A. M. Balazuc, H. Kiefer-Biasizzo, J. R. Lapa e Silva, G. Milon, G. Marchal, and B. B. Vargaftig. 2003. Dendritic cells recruited to the lung shortly after intranasal delivery of Mycobacterium bovis BCG drive the primary immune response towards a type 1 cytokine production. Immunology108:352-364.
    OpenUrlCrossRefPubMedWeb of Science
  25. 25.↵
    Leemans, J. C., N. P. Juffermans, S. Florquin, N. van Rooijen, M. J. Vervoordeldonk, A. Verbon, S. J. van Deventer, and T. van der Poll. 2001. Depletion of alveolar macrophages exerts protective effects in pulmonary tuberculosis in mice. J. Immunol.166:4604-4611.
    OpenUrlAbstract/FREE Full Text
  26. 26.↵
    Legge, K. L., and T. J. Braciale. 2003. Accelerated migration of respiratory dendritic cells to the regional lymph nodes is limited to the early phase of pulmonary infection. Immunity18:265-277.
    OpenUrlCrossRefPubMedWeb of Science
  27. 27.↵
    LeVine, A. M., K. E. Kurak, M. D. Bruno, J. M. Stark, J. A. Whitsett, and T. R. Korfhagen. 1998. Surfactant protein-A-deficient mice are susceptible to Pseudomonas aeruginosa infection. Am. J. Respir. Cell Mol. Biol.19:700-708.
    OpenUrlCrossRefPubMedWeb of Science
  28. 28.↵
    LeVine, A. M., J. A. Reed, K. E. Kurak, E. Cianciolo, and J. A. Whitsett. 1999. GM-CSF deficient mice are susceptible to pulmonary group B streptococcal infection. J. Clin. Investig.103:563-569.
    OpenUrlCrossRefPubMedWeb of Science
  29. 29.↵
    Maus, U., S. Herold, H. Muth, R. Maus, L. Ermert, M. Ermert, N. Weissmann, S. Rosseau, W. Seeger, F. Grimminger, and J. Lohmeyer. 2001. Monocytes recruited into the alveolar air space of mice show a monocytic phenotype but upregulate CD14. Am. J. Physiol. Lung Cell. Mol. Physiol.280:L58-L68.
    OpenUrlPubMedWeb of Science
  30. 30.↵
    Means, T. K., S. Wang, E. Lien, A. Yoshimura, D. T. Golenbock, and M. J. Fenton. 1999. Human toll-like receptors mediate cellular activation by Mycobacterium tuberculosis.J. Immunol.163:3920-3927.
    OpenUrlAbstract/FREE Full Text
  31. 31.↵
    Noss, E. H., C. V. Harding, and W. H. Boom. 2000. Mycobacterium tuberculosis inhibits MHC class II antigen processing in murine bone marrow macrophages. Cell. Immunol.201:63-74.
    OpenUrlCrossRefPubMed
  32. 32.↵
    Noss, E. H., R. K. Pai, T. J. Sellati, J. D. Radolf, J. Belisle, D. T. Golenbock, W. H. Boom, and C. V. Harding. 2001. Toll-like receptor 2-dependent inhibition of macrophage class II MHC expression and antigen processing by 19-kDa lipoprotein of Mycobacterium tuberculosis. J. Immunol.167:910-918.
    OpenUrlAbstract/FREE Full Text
  33. 33.↵
    Orme, I. M., P. Andersen, and W. H. Boom. 1993. T cell response to Mycobacterium tuberculosis.J. Infect. Dis.167:1481-1497.
    OpenUrlCrossRefPubMedWeb of Science
  34. 34.↵
    Pai, R. K., M. Convery, T. A. Hamilton, W. H. Boom, and C. V. Harding. 2003. Inhibition of IFN-γ-induced class II transactivator expression by a 19-kDa lipoprotein from Mycobacterium tuberculosis: a potential mechanism for immune evasion. J. Immunol.171:175-184.
    OpenUrlAbstract/FREE Full Text
  35. 35.↵
    Paine, R., III, S. B. Morris, H. Jin, S. E. Wilcoxen, S. M. Phare, B. B. Moore, M. J. Coffey, and G. B. Toews. 2001. Impaired functional activity of alveolar macrophages from GM-CSF-deficient mice. Am. J. Physiol. Lung Cell. Mol. Physiol.281:L1210-L1218.
    OpenUrlPubMedWeb of Science
  36. 36.↵
    Pasula, R., J. R. Wright, D. L. Kachel, and W. J. Martin II. 1999. Surfactant protein A suppresses reactive nitrogen intermediates by alveolar macrophages in response to Mycobacterium tuberculosis. J. Clin. Investig.103:483-490.
    OpenUrlCrossRefPubMedWeb of Science
  37. 37.↵
    Pearl, J. E., B. Saunders, S. Ehlers, I. M. Orme, and A. M. Cooper. 2001. Inflammation and lymphocyte activation during mycobacterial infection in the interferon-gamma-deficient mouse. Cell. Immunol.211:43-50.
    OpenUrlCrossRefPubMedWeb of Science
  38. 38.↵
    Rich, E. A., M. Torres, E. Sada, C. K. Finegan, B. D. Hamilton, and Z. Toossi. 1997. Mycobacterium tuberculosis (MTB)-stimulated production of nitric oxide by human alveolar macrophages and relationship of nitric oxide production to growth inhibition of MTB. Tuber. Lung Dis.78:247-255.
    OpenUrlCrossRefPubMedWeb of Science
  39. 39.↵
    Sadek, M. I., E. Sada, Z. Toossi, S. K. Schwander, and E. A. Rich. 1998. Chemokines induced by infection of mononuclear phagocytes with mycobacteria and present in lung alveoli during active pulmonary tuberculosis. Am. J. Respir. Cell Mol. Biol.19:513-521.
    OpenUrlCrossRefPubMedWeb of Science
  40. 40.↵
    Schauble, T. L., W. H. Boom, C. K. Finegan, and E. A. Rich. 1993. Characterization of suppressor function of human alveolar macrophages for T lymphocyte responses to phytohemagglutinin: cellular selectivity, reversibility and early events in T cell activation. Am. J. Respir. Cell Mol. Biol.8:89-97.
    OpenUrlCrossRefPubMedWeb of Science
  41. 41.↵
    Schneeberger, E. E., M. DeFerrari, M. J. Skoskiewicz, P. S. Russell, and R. B. Colvin. 1986. Induction of MHC-determined antigens in the lung by interferon-γ. Lab. Investig.55:138-144.
    OpenUrlPubMed
  42. 42.↵
    Schwander, S. K., M. Torres, E. Sada, C. Carranza, E. Ramos, M. Tary-Lehmann, R. S. Wallis, J. Sierra, and E. A. Rich. 1998. Enhanced responses to Mycobacterium tuberculosis antigens by human alveolar lymphocytes during active pulmonary tuberculosis. J. Infect. Dis.178:1434-1445.
    OpenUrlCrossRefPubMedWeb of Science
  43. 43.↵
    Soltys, J., T. Bonfield, J. Chmiel, and M. Berger. 2002. Functional IL-10 deficiency in the lung of cystic fibrosis (cftr−/−) and IL-10 knockout mice causes increased expression and function of B7 costimulatory molecules on alveolar macrophages. J. Immunol.168:1903-1910.
    OpenUrlAbstract/FREE Full Text
  44. 44.↵
    Stokes, R. W., L. M. Thorson, and D. P. Speert. 1998. Nonopsonic and opsonic association of Mycobacterium tuberculosis with resident alveolar macrophages is inefficient. J. Immunol.160:5514-5521.
    OpenUrlAbstract/FREE Full Text
  45. 45.↵
    Strieter, R. M., S. W. Chensue, M. A. Basha, T. J. Standiford, J. P. Lynch, M. Baggiolini, and S. L. Kunkel. 1990. Human alveolar macrophage gene expression of interleukin-8 by tumor necrosis factor-α, lipopolysaccharide, and interleukin-1-β. Am. J. Respir. Cell Mol. Biol.2:321-326.
    OpenUrlCrossRefPubMedWeb of Science
  46. 46.↵
    Takeda, K., T. Kaisho, and S. Akira. 2003. Toll-like receptors. Annu. Rev. Immunol.21:335-376.
    OpenUrlCrossRefPubMedWeb of Science
  47. 47.↵
    Ting, L. M., A. C. Kim, A. Cattamanchi, and J. D. Ernst. 1999. Mycobacterium tuberculosis inhibits IFN-γ transcriptional responses without inhibiting activation of STAT1. J. Immunol.163:3898-3906.
    OpenUrlAbstract/FREE Full Text
  48. 48.↵
    Toews, G. B., W. C. Vial, M. M. Dunn, P. Guzzetta, G. Nunez, P. Stastny, and M. F. Lipscomb. 1984. The accessory cell function of human alveolar macrophages in specific T cell proliferation. J. Immunol.132:181-186.
    OpenUrlAbstract
  49. 49.↵
    Toossi, Z., P. Gogate, H. Shiratsuchi, T. Young, and J. J. Ellner. 1995. Enhanced production of TGF-β by blood monocytes from patients with active tuberculosis and presence of TGF-β in tuberculous granulomatous lung lesions. J. Immunol.154:465-473.
    OpenUrlAbstract
  50. 50.↵
    Tsao, T. C., J. Hong, C. Huang, P. Yang, S. K. Liao, and K. S. Chang. 1999. Increased TNF-alpha, IL-1 beta and IL-6 levels in the bronchoalveolar lavage fluid with the upregulation of their mRNA in macrophages lavaged from patients with active pulmonary tuberculosis. Tuber. Lung Dis.79:279-285.
    OpenUrlCrossRefPubMed
  51. 51.↵
    Underhill, D. M., A. Ozinsky, K. D. Smith, and A. Aderem. 1999. Toll-like receptor-2 mediates mycobacteria-induced proinflammatory signaling in macrophages. Proc. Natl. Acad. Sci. USA96:14459-14463.
    OpenUrlAbstract/FREE Full Text
  52. 52.↵
    van Crevel, R., T. H. M. Ottenhoff, and J. W. M. van der Meer. 2002. Innate immunity to Mycobacterium tuberculosis.Clin. Microbiol. Rev.15:294-309.
    OpenUrlAbstract/FREE Full Text
  53. 53.↵
    Zhang, P., W. R. Summer, G. J. Bagby, and S. Nelson. 2000. Innate immunity and pulmonary host defense. Immunol. Rev.173:39-51.
    OpenUrlCrossRefPubMedWeb of Science
PreviousNext
Back to top
Download PDF
Citation Tools
Inhibition of Major Histocompatibility Complex II Expression and Antigen Processing in Murine Alveolar Macrophages by Mycobacterium bovis BCG and the 19-Kilodalton Mycobacterial Lipoprotein
Scott A. Fulton, Scott M. Reba, Rish K. Pai, Meghan Pennini, Martha Torres, Clifford V. Harding, W. Henry Boom
Infection and Immunity Mar 2004, 72 (4) 2101-2110; DOI: 10.1128/IAI.72.4.2101-2110.2004

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Print

Alerts
Sign In to Email Alerts with your Email Address
Email

Thank you for sharing this Infection and Immunity article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Inhibition of Major Histocompatibility Complex II Expression and Antigen Processing in Murine Alveolar Macrophages by Mycobacterium bovis BCG and the 19-Kilodalton Mycobacterial Lipoprotein
(Your Name) has forwarded a page to you from Infection and Immunity
(Your Name) thought you would be interested in this article in Infection and Immunity.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
Inhibition of Major Histocompatibility Complex II Expression and Antigen Processing in Murine Alveolar Macrophages by Mycobacterium bovis BCG and the 19-Kilodalton Mycobacterial Lipoprotein
Scott A. Fulton, Scott M. Reba, Rish K. Pai, Meghan Pennini, Martha Torres, Clifford V. Harding, W. Henry Boom
Infection and Immunity Mar 2004, 72 (4) 2101-2110; DOI: 10.1128/IAI.72.4.2101-2110.2004
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Top
  • Article
    • ABSTRACT
    • MATERIALS AND METHODS
    • RESULTS
    • DISCUSSION
    • ACKNOWLEDGMENTS
    • FOOTNOTES
    • REFERENCES
  • Figures & Data
  • Info & Metrics
  • PDF

KEYWORDS

antigen presentation
Antigen-Presenting Cells
Histocompatibility Antigens Class II
lipoproteins
Macrophages, Alveolar
Mycobacterium bovis

Related Articles

Cited By...

About

  • About IAI
  • Editor in Chief
  • Editorial Board
  • Policies
  • For Reviewers
  • For the Media
  • For Librarians
  • For Advertisers
  • Alerts
  • RSS
  • FAQ
  • Permissions
  • Journal Announcements

Authors

  • ASM Author Center
  • Submit a Manuscript
  • Article Types
  • Ethics
  • Contact Us

Follow #IAIjournal

@ASMicrobiology

       

ASM Journals

ASM journals are the most prominent publications in the field, delivering up-to-date and authoritative coverage of both basic and clinical microbiology.

About ASM | Contact Us | Press Room

 

ASM is a member of

Scientific Society Publisher Alliance

 

American Society for Microbiology
1752 N St. NW
Washington, DC 20036
Phone: (202) 737-3600

Copyright © 2021 American Society for Microbiology | Privacy Policy | Website feedback

Print ISSN: 0019-9567; Online ISSN: 1098-5522