Skip to main content

Main menu

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons

User menu

  • Subscribe
  • My alerts
  • Log in
  • Log out

Search

  • Advanced search
The Journal of Immunology
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons
  • Subscribe
  • My alerts
  • Log in
  • Log out
The Journal of Immunology

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Follow The Journal of Immunology on Twitter
  • Follow The Journal of Immunology on RSS

Reactive Oxygen Species and 12/15-Lipoxygenase Contribute to the Antiproliferative Capacity of Alternatively Activated Myeloid Cells Elicited during Helminth Infection

Lea Brys, Alain Beschin, Geert Raes, Gholamreza Hassanzadeh Ghassabeh, Wim Noël, Jef Brandt, Frank Brombacher and Patrick De Baetselier
J Immunol May 15, 2005, 174 (10) 6095-6104; DOI: https://doi.org/10.4049/jimmunol.174.10.6095
Lea Brys
*Department of Cellular and Molecular Interactions, Vlaams Interuniversitair Instituut voor Biotechnologie, Vrije Universiteit Brussel, Brussels, Belgium; †Diergeneeskunde, Instituut voor Tropische Geneeskunde, Antwerpen, Belgium; and ‡Health Sciences Faculty, University of Cape Town, Cape Town, South Africa
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Alain Beschin
*Department of Cellular and Molecular Interactions, Vlaams Interuniversitair Instituut voor Biotechnologie, Vrije Universiteit Brussel, Brussels, Belgium; †Diergeneeskunde, Instituut voor Tropische Geneeskunde, Antwerpen, Belgium; and ‡Health Sciences Faculty, University of Cape Town, Cape Town, South Africa
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Geert Raes
*Department of Cellular and Molecular Interactions, Vlaams Interuniversitair Instituut voor Biotechnologie, Vrije Universiteit Brussel, Brussels, Belgium; †Diergeneeskunde, Instituut voor Tropische Geneeskunde, Antwerpen, Belgium; and ‡Health Sciences Faculty, University of Cape Town, Cape Town, South Africa
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Gholamreza Hassanzadeh Ghassabeh
*Department of Cellular and Molecular Interactions, Vlaams Interuniversitair Instituut voor Biotechnologie, Vrije Universiteit Brussel, Brussels, Belgium; †Diergeneeskunde, Instituut voor Tropische Geneeskunde, Antwerpen, Belgium; and ‡Health Sciences Faculty, University of Cape Town, Cape Town, South Africa
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Wim Noël
*Department of Cellular and Molecular Interactions, Vlaams Interuniversitair Instituut voor Biotechnologie, Vrije Universiteit Brussel, Brussels, Belgium; †Diergeneeskunde, Instituut voor Tropische Geneeskunde, Antwerpen, Belgium; and ‡Health Sciences Faculty, University of Cape Town, Cape Town, South Africa
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jef Brandt
*Department of Cellular and Molecular Interactions, Vlaams Interuniversitair Instituut voor Biotechnologie, Vrije Universiteit Brussel, Brussels, Belgium; †Diergeneeskunde, Instituut voor Tropische Geneeskunde, Antwerpen, Belgium; and ‡Health Sciences Faculty, University of Cape Town, Cape Town, South Africa
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Frank Brombacher
*Department of Cellular and Molecular Interactions, Vlaams Interuniversitair Instituut voor Biotechnologie, Vrije Universiteit Brussel, Brussels, Belgium; †Diergeneeskunde, Instituut voor Tropische Geneeskunde, Antwerpen, Belgium; and ‡Health Sciences Faculty, University of Cape Town, Cape Town, South Africa
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Patrick De Baetselier
*Department of Cellular and Molecular Interactions, Vlaams Interuniversitair Instituut voor Biotechnologie, Vrije Universiteit Brussel, Brussels, Belgium; †Diergeneeskunde, Instituut voor Tropische Geneeskunde, Antwerpen, Belgium; and ‡Health Sciences Faculty, University of Cape Town, Cape Town, South Africa
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Understanding the role of CD11b+GR-1+ myeloid suppressor cells in the immune suppression and immunoregulation associated with a variety of diseases may provide therapeutic opportunities. In this article, we show, in a model of helminth infection, that CD11b+GR-1+ myeloid suppressor cells but not CD11b+F4/80high mature macrophages expanded in the peritoneal cavity of BALB/c mice implanted with Taenia crassiceps. Peritoneal cell populations from early stage-infected animals impaired T cell proliferation by secreting NO. Yet, they lost their ability to secrete NO in the late stage of infection. Concomitantly, their capacity to exert arginase activity and to express mRNAs coding for FIZZ1 (found in inflammatory zone 1), Ym, and macrophage galactose-type C-type lectin increased. Furthermore, cells from early stage-infected mice triggered T cells to secrete IFN-γ and IL-4, whereas in the late stage of infection, they only induced IL-4 production. These data suggest that CD11b+GR-1+ myeloid suppressor cells displaying an alternative activation phenotype emerged gradually as T. crassiceps infection progressed. Corroborating the alternative activation status in the late stage of infection, the suppressive activity relied on arginase activity, which facilitated the production of reactive oxygen species including H2O2 and superoxide. We also document that the suppressive activity of alternative myeloid suppressor cells depended on 12/15-lipoxygenase activation generating lipid mediators, which triggered peroxisome proliferator-activated receptor-γ. IL-4 and IL-13 signaling contributed to the expansion of myeloid suppressor cells in the peritoneal cavity of T. crassiceps-infected animals and to their antiproliferative activity by allowing arginase and 12/15-lipoxygenase gene expression.

About 3 decades ago, non-B, non-T accessory cells hampering immune responses without prior activation were described as natural suppressor cells (1, 2). Recent evidence suggests that these cells are related closely to a population of suppressive cells that express surface markers common to the macrophage (CD11b) and the granulocyte (GR-1) lineage (3, 4).

Myeloid suppressor CD11b+GR-1+ cells represent an intrinsic part of myeloid cell differentiation in the bone marrow (5). They are present in healthy individuals, where their main function in vivo is probably to restrain overwhelming immune responses. However, myeloid suppressor cells (MSC) 3 accumulate in a variety of immune conditions associated with depression of adaptive immune responses. As such, they may constitute a barrier to the therapy of various diseases. Conversely, they may represent therapeutic opportunities to moderate unwanted immune responses, occurring, for instance, during autoimmune diseases or allograft rejection. Consequently, the mechanisms by which MSC influence immune responses were addressed critically in the last years.

MSC interfere with the activation and proliferation of B cells and CD8+ and CD4+ T cells. They can also cause cell death by apoptosis (4, 6). Two major populations of MSC were characterized, based on their differential l-arginine metabolism and the cytokine environment in which they develop. Classical MSC, elicited by IFN-γ released by activated Th1 cells, metabolize l-arginine via inducible NO synthase (iNOS), producing NO and l-citrulline. They were described in systems that include strong immunization procedures (7, 8, 9, 10), chemotherapy with cyclophosphamide (3, 11), tumor growth (3, 11, 12, 13, 14, 15, 16), as well as during bacterial (17, 18) and American trypanosome (19) infections. Classical MSC inhibit T cell proliferation via their NO secretion, which is elicited by IFN-γ and a yet unknown contact between MSC and T cells. Alternative MSC act on l-arginine via arginase activity to produce urea and l-ornithine. To date, they were found in tumor-bearing mice, being elicited by IL-4 or IL-13 released by Th2 cells or CD1d-restricted NKT cells (20, 21, 22, 23). Their suppressive activity depends on a cell contact with T cells, possibly via adhesion molecules, and results from the depletion of l-arginine through arginase activity, hereby favoring the iNOS reductase activity leading to increased production of reactive oxygen species (ROS).

In models where the nematode Brugia malayi or the cestode Taenia crassiceps is implanted in the peritoneal cavity of mice, infection is characterized by a bias toward a Th2 immune response, alternative activation of myeloid cells, and impaired proliferation of immune cells. However, the nature and mechanisms of action of suppressive cells remain poorly characterized. Indeed, although the suppressive activity was assigned to F4/80+ macrophage-like cells in T. crassiceps-infected mice, both F4/80− and F4/80+ suppressive cells were postulated to be induced during B. malayi infection (24, 25, 26, 27, 28). In addition, NO-independent suppressive mechanisms were evidenced in the late stage of infection with helminths (24, 25, 26, 27, 28, 29), but whether this is valid throughout the infection period is not documented.

In this article, we attempted to get better insight in the phenotype as well as the mechanisms underlying the antiproliferative potential of cells elicited in the peritoneal cavity of BALB/c mice implanted with T. crassiceps at different time points postinfection. We report the gradual emergence in the course of infection of alternative CD11b+GR-1+ MSC, out of a population that in the early stage of infection displays the features of classical and alternative activation. The change in the activation status as the infection progresses is paralleled by a switch from NO-dependent to ROS- and/or 12/15-lipoxygenase (12/15-LOX)-dependent suppressive activity. Finally, our data also reveal that the development of this type of MSC depends on IL-4 and/or IL-13.

Materials and Methods

Infection

Female wild-type (Harlan), IL-4−/− (30), or IL-4Rα−/− (31) BALB/c were inoculated i.p. with 10 nonbudding Toi strain T. crassiceps metacestodes as described previously (27) so that, at the time of the experiment, age-matched animals from both the early (<4 wk) and late (>6 wk) stage of infection were available.

Isolation of peritoneal cell populations

Resident peritoneal cells were collected in 0.34 M sucrose and washed in complete medium (RPMI 1640 medium supplemented with 10% heat-inactivated FCS, 5 × 10−5 M 2-ME, 2 mM l-glutamine, 100 IU/ml penicillin, 100 μg/ml streptomycin, and 0.1 mM nonessential amino acids; all from Invitrogen Life Technologies). Adherent cells were recovered after plastic adherence as described previously (27). Collected cells had a viability >95%, as determined by trypan blue exclusion, and were CD11b+ for ∼90%, as determined by flow cytometry analysis (data not shown).

FACS staining

Cells were stained for 20 min at 4°C using conventional protocols. Cells were incubated with anti-FcγR Ab (clone 2.4G2) before adding (1 μg/106 cells): FITC/PE-conjugated anti-CD11b (clone M1/70), PE-conjugated anti-GR-1 (Ly6C/G, clone RB6-8C5), PE-conjugated anti-CD3 (clone 145-2C11), PE-conjugated anti-CD19 (clone 1D3), PE-conjugated anti-F4/80 (clone CI:A3-1), or PE-conjugated irrelevant control Abs (clone G155-178). All Abs were purchased from BD Biosciences, with the exception of anti-F4/80 (Serotec). Cells (104) were analyzed on a FACSVantage SE flow cytometer (BD Biosciences) using CellQuest program.

Quantification of NO2 and arginase activity

Peritoneal cells (2 × 105/200 μl) were cultured in humidified atmosphere containing 5% CO2 in complete medium in the absence or the presence of LPS (0.1 μg/ml, Escherichia coli O55:B5) for 48 h at 37°C. NO in culture supernatants was estimated by quantifying NO2 using Greiss reagent as described previously (27). Arginase activity was measured in peritoneal cell lysates as described previously (32).

RNA extraction and quantitative RT-PCR analysis

One microgram of total RNA prepared using TRIzol reagent (Invitrogen Life Technologies) was reverse-transcribed using oligo(dT) and SuperScript II reverse transcriptase (Invitrogen Life Technologies) following the manufacturer’s recommendations. Quantitative real-time PCR was performed in a Bio-Rad iCycler, with Bio-Rad iQ SYBR Green Supermix using as primers: ribosomal protein S12 sense (5′-CCTCGATGACATCCTTGGCCTGAG-3′) and antisense (5′-GGAAGGCATAGCTGCTGGAGGTGT-3′); arginase 1 sense (5′-ATGGAAGAGACCTTCAGCTAC-3′) and antisense (5′-GCTGTCTTCCCAAGAGTTGGG-3′); found in inflammatory zone 1 (FIZZ1) sense (5′-TCCCAGTGAATACTGATGAGA-3′) and antisense (5′-CCACTCTGGATCTCCCAAGA-3′); Ym sense (5′-GGGCATACCTTTATCCTGAG-3′) and antisense (5′-CCACTG AAGTCATCCATGTC-3′); MGL2 sense (5′-GATAACTGGCATGGACATATG-3′) and antisense (5′-TTTCTAATCACCATAACACATTC-3′); and 12/15-LOX sense (5′-CAGCTGGATTGGTTCTACTG-3′) and antisense (5′-CTCTGAACTTCTTCAGCACAG-3′). For all primers, each PCR cycle consisted of 1-min denaturation at 94°C, 45 s annealing at 55°C, and 1-min extension at 72°C. Gene expression was normalized using ribosomal protein S12 as housekeeping gene. Similar results were obtained using other housekeeping genes.

Proliferative assay

Single cell suspensions of mesenteric, axillary, and inguinal lymph nodes from noninfected mice (2 × 105) were cocultured with adherent peritoneal cells (at a ratio of 5%) and activated with Con A (2.5 μg/ml; Sigma-Aldrich) or anti-CD3 Ab (clone 145-2C11, 1 μg/ml; BD Biosciences) in 200 μl of complete medium. When required, the following inhibitors were used: NG-monomethyl l-arginine (l-NMMA) (1000 μΜ; Sigma-Aldrich), N(omega)-hydroxy-nor-l-arginine (nor-NOHA) (250 μM; Bachem), superoxide dismutase (SOD) (200 U/ml; Sigma-Aldrich), catalase (300 U/ml; Sigma), nordihydroguaiaretic acid (NDGA) (10 μM; Sigma-Aldrich), GW9662 (10 μM; Sigma-Aldrich), and indomethacin (10 μg/ml). Stock solutions of NDGA, GW9662, and indomethacin were made in DMSO. Therefore, a corresponding volume of DMSO was added to cocultures performed in the absence of these inhibitors. After 24 h at 37°C in a humidified atmosphere containing 5% CO2, cells were pulsed for 18 h with 1 μCi of [methyl-3H]-thymidine ([3H]Thy) (TRA310; Amersham Biosciences) and harvested onto glass fiber filters, and the incorporated radioactivity was measured in a liquid scintillator counter (1450 microbeta; Wallac). Results were expressed as the [3H]Thy incorporation from triplicate cultures subtracting background values from nonstimulated cells. Proliferative responses (mean cpm ± SD of three mice tested individually) were compared in cocultures containing peritoneal cells from infected and noninfected mice.

Cytokine quantification

Cytokines were quantified using sandwich ELISAs for IFN-γ and IL-4 performed as recommended by the supplier (BD Biosciences). Maximal levels of cytokines observed after 3 days (IFN-γ) or 1 day (IL-4) of culture are reported.

Statistical analysis

All comparisons were tested for statistical significance (p < 0.05) via the unpaired t test, using GraphPad Prism 3.0 software.

Results

CD11b+GR-1+ myeloid cells are induced on T. crassiceps infection

Implantation of T. crassiceps in the peritoneal cavity of BALB/c mice caused a massive accumulation of cells in this compartment (Fig. 1⇓). The cell number gradually increased, reaching a maximum 3–4 wk postinfection of 63.5 ± 11 × 106 cells, as compared with 3.6 ± 1.8 × 106 cells in noninfected animals. The cell number then decreased and stabilized at ∼20 × 106 between the 6th and 12th wk of infection, time at which the experiment was stopped.

           FIGURE 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1.

T. crassiceps infection causes CD11b+ cell accumulation in the peritoneal cavity. At indicated time points postinfection, the total cell number (a) recovered from the peritoneal cavity of three infected mice was determined using a hemocytometer and compared with the cell number in two noninfected animals. In parallel, percentages of CD11b+ (b), CD3+ (c), and CD19+ cells (d) in the total cell population were determined by flow cytometry. At each time point postinfection investigated, the mean ± SD of infected (▪) and noninfected (▴) mice was compared. One representative of three independent experiments is shown. ∗, p < 0.05 higher comparing infected vs noninfected mice. #, p < 0.05 lower comparing infected vs noninfected mice.

The modulation of the total cell number in the peritoneal cavity of T. crassiceps-infected animals was paralleled by a gradual increase in the percentage of CD11b+ myeloid cells, from 38 ± 5% in noninfected mice to ∼75% in late stage-infected animals (Fig. 1⇑). Initially, the percentage of CD3+ T cells increased, reaching a maximum of 42 ± 5% 1 wk after T. crassiceps implantation, as compared with the 24 ± 8% observed in noninfected mice, but became lower than in noninfected animals from the fourth week of infection. The percentage of CD19+ B cells dropped very rapidly, from 38 ± 8% of the total cell population in noninfected mice to <5% in the late stage-infected mice.

Further characterization of the phenotype of peritoneal CD11b+ cells revealed that the percentage of mature macrophages, coexpressing high levels of CD11b and F4/80 (33), tended to decrease upon T. crassiceps infection, in particular from 4–5 wk postinfection (Fig. 2⇓). In contrast, the percentage of CD11blowF4/80low cells increased (see Fig. 4⇓). In addition, CD11blow cells that emerged gradually in the peritoneal cavity of infected mice coexpressed the myeloid epitope GR-1 (Fig. 2⇓) at low or high levels (see Figs. 4⇓ and 8⇓). These CD11blowGR-1low and CD11blowGR-1high populations displayed low and high scatters, respectively (data not shown), likely reflecting the expansion of immature myeloid cells and granulocytes (33) in T. crassiceps-infected mice. May-Grunwald Giemsa staining of the GR-1low and GR-1high cells sorted from MACS-purified CD11b+ peritoneal cells confirmed their nature as macrophage-like cells and granulocytes, respectively (Fig. 2⇓). A CD11blowGR-1− population decreasing gradually in the course of infection (see Figs. 4⇓ and 8⇓) possibly represented B cells (34).

           FIGURE 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2.

CD11b+GR-1+ cells expand in the peritoneal cavity of T. crassiceps-infected mice. At indicated time points postinfection, percentages of CD11blowGR-1+ (A) and CD11bhighF4/80high cells (B) in the total cell population were determined by flow cytometry, as described in Fig. 1⇑, using the gates depicted in Fig. 4⇓ (▪, infected mice; ▴, noninfected mice). ∗, p < 0.05 higher comparing infected vs noninfected mice. CD11b+GR-1low (C) and CD11b+GR-1high (D) cells were sorted from the peritoneal cavity of infected mice (2 wk postinfection) and stained with May-Grunwald Giemsa (×400).

Collectively, although the percentage of T and B cells decreased gradually, the proportion of CD11b+ cells expanded up to two times in the peritoneal cavity of Taenia-infected animals as the disease progressed. Concomitantly, the percentage of CD11bhighF4/80high mature macrophages tended to decrease, whereas the percentage of CD11blow cells likely consisting of immature myeloid cells (GR-1low) and granulocytes (GR-1high) increased up to ten times.

Alternative myeloid cells emerge gradually in the course of T. crassiceps infection

T. crassiceps-infected mice mount a mixed Th1/Th2 cytokine response in the early stage of infection that shifts to a more polarized Th2 cytokine response in the late phase of the disease due to a decrease in IFN-γ production (28, 35, 36). Considering that Th1 and Th2 cytokines antagonistically regulate the development of classical and alternative myeloid cells, the NO/arginase balance was investigated in T. crassiceps-infected mice. As reported previously (27), peritoneal cells from infected mice were sensitized to secrete higher amounts of NO than equivalent cells from noninfected mice only in the early stage of infection, in particular upon LPS stimulation (Fig. 3⇓). In contrast, arginase activity was evidenced from the first week of infection and increased as the infection progressed to reach maximal values ∼4 wk postinfection. NO secretion as well as arginase activity were confined to the adherent fraction of peritoneal cells (data not shown). Thus, we focused on this cell population in the work described below. Data from representative experiments performed in the early (<4 wk) and late (>6 wk) stage of infection are illustrated.

           FIGURE 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 3.

NO production and arginase activity in the peritoneal cavity of T. crassiceps-infected mice display reciprocal kinetics. At indicated time points postinfection, NO production was quantified in supernatants of peritoneal cells from three infected mice and two noninfected animals cultured in the absence (a) or the presence of LPS (b). In parallel, arginase activity was determined in peritoneal cell lysates (c). At each time point postinfection investigated, the mean ± SD of infected (▪) and noninfected animals (▴) was compared. One representative of three independent experiments is shown. ∗, p < 0.05 higher comparing infected vs noninfected mice. #, p < 0.05 lower comparing infected vs noninfected mice.

In the adherent peritoneal cell fraction, a gradual increase in CD11blow cells coexpressing GR-1low and/or F4/80low was observed in the course of T. crassiceps infection as compared with noninfected mice (Fig. 4⇓ and Table I⇓). The percentage of CD11blowGR-1high cells also increased but only in the early stage of infection. The percentage of mature macrophages, i.e., CD11bhigh cells expressing F4/80high and being GR-1−, did not differ significantly in the adherent cell fraction of infected and noninfected mice at the times investigated postinfection, and, if anything, it tended to be lower in infected animals. Finally, the percentage of adherent CD11blow cells being GR-1− or F4/80− was lower in infected than in noninfected mice. Thus, mainly CD11blowGR-1low (F4/80low) immature myeloid cells were enriched in the adherent fraction of peritoneal cells from T. crassiceps-infected mice as compared with noninfected animals.

           FIGURE 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 4.

Adherent peritoneal cells from T. crassiceps-infected mice are enriched in CD11blowGR-1+ cells. Adherent cell populations collected in the early (<4 wk) and late (>6 wk) stage of infection were stained with FITC-labeled anti-CD11b and PE-labeled anti-GR-1 or PE-labeled anti-F4/80 Abs. Simultaneously, cells from three noninfected mice were pooled and submitted to the same staining procedure. The percentage of gated positive cells was determined. Profiles are representative of one pool of cells from noninfected mice and of cells from one infected individual of seven investigated at each stage of infection in three independent experiments.

View this table:
  • View inline
  • View popup
Table I.

Percentage of GR-1+ and F4/80+ cells within adherent CD11b+ peritoneal cells collected in wild-type mice in the early and late stage of T. crassicepsinfectiona

Besides arginase, the expression of the genes coding for FIZZ1, Ym, and the macrophage galactose-type C-type lectin (MGL) family (37, 38, 39) is up-regulated in alternatively activated macrophages elicited during parasite infections. Expression levels of these genes were thus analyzed by real-time PCR to further address the activation status of adherent peritoneal cells in T. crassiceps-infected mice. As compared with noninfected mice, FIZZ1, Ym, and MGL mRNA expression gradually increased in the course of infection (Fig. 5⇓). The up-regulation of the expression of these genes occurred as early as 4 days postinfection (data not shown).

           FIGURE 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 5.

Modulation of FIZZ1, Ym, and MGL mRNA expression in adherent peritoneal cells from T. crassiceps-infected mice. Gene expression of FIZZ1 (a), Ym (b), and MGL (c) was determined via quantitative real-time PCR and normalized for the housekeeping gene ribosomal protein S12 in adherent peritoneal cells from infected mice collected in the early (< 4 wk) and late stage of infection (>6 wk). The fold induction of the gene expression (mean ± SD of three individual animals) in infected as compared with noninfected mice is shown for one representative of three independent experiments. ∗, p < 0.05 higher comparing infected vs noninfected mice.

In agreement with a previous report (27), adherent peritoneal cells from early stage-infected mice triggered the production of both IFN-γ and IL-4 when cocultured with lymph node cells from noninfected mice activated with Con A, whereas cells from late stage-infected animals induced only the release of IL-4 (Table II⇓).

View this table:
  • View inline
  • View popup
Table II.

IFN-γ and IL-4 production in supernatants of Con A-activated lymph node cells from noninfected mice cocultured with adherent peritoneal cells from T. crassiceps-infected animalsa

Collectively, these data suggest that CD11blowGR-1+F4/80low myeloid cells displaying characteristics of classical and alternative activation status developed in the early stage of T. crassiceps infection. Yet, a polarization toward alternative myeloid cells occurred gradually as the infection progressed. The change in the activation status further correlated with the capacity of myeloid cells from early or late stage-infected mice to elicit a mixed Th1/Th2 or a Th2 cytokine profile, respectively.

iNOS and arginase contribute differentially to the suppressive activity of classical and alternative myeloid cells

Previous studies have documented the existence of a profound T cell unresponsiveness in T. crassiceps-infected mice (35, 40, 41, 42). Because classical as well as alternative CD11b+GR-1+ myeloid cells have been implicated in immunosuppression, the ability of adherent cells from the peritoneal cavity of mice implanted with T. crassiceps to impair the proliferation of T cell from noninfected animals was investigated in the presence of the NO inhibitor l-NMMA and the arginase inhibitor nor-NOHA (Fig. 6⇓).

           FIGURE 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 6.

NO and arginase contribute to the antiproliferative capacity of adherent peritoneal cells from T. crassiceps-infected mice. Adherent peritoneal cells from three infected and three noninfected mice were cocultured with lymph node cells from noninfected mice activated with Con A in the absence or presence of NO (l-NMMA) and/or arginase (nor-NOHA) inhibitor. Proliferation was determined by [3H]Thy incorporation. The mean ± SD of infected and noninfected mice, of one representative of three independent experiments performed in the early (<4 wk) (a) and late (>6 wk) (b) stage of infection, is shown. ∗, p < 0.05 lower comparing infected vs noninfected mice. #, p < 0.05 higher comparing infected mice in the presence vs the absence of inhibitor.

In the early stage of infection, l-NMMA nearly completely reversed the suppressive activity of cells from T. crassiceps-infected mice on Con A-induced T cell proliferation (by 86 ± 14%). At this stage, nor-NOHA could not restore proliferation, despite the fact that cells from infected mice exerted significant arginase activity (Fig. 3⇑).

In the late stage of infection, the suppressive activity exerted by adherent peritoneal cells from Taenia-infected mice on Con A-induced T cell proliferation was not affected by l-NMMA and was marginally reduced by nor-NOHA (by 28 ± 7%) (Fig. 6⇑). Noteworthy at this stage, the combination of the two inhibitors almost fully restored the ability of T cells to proliferate (by 88 ± 12%).

Recent studies have shown that alternative MSC exert their suppressive activity through oxidative stress and that arginase activity is the major contributor to the production of ROS (23, 32). Therefore, the potential role of ROS in the inhibition of T cell proliferation triggered by adherent alternative myeloid cells elicited in the late stage of T. crassiceps infection was evaluated (Fig. 7⇓). SOD and catalase were found to restore partially but significantly the proliferation of T cell induced by Con A (by 41 ± 10% and 45 ± 10%, respectively). Hence, superoxide and H2O2 take part in the suppressive activity of alternative MSC elicited during T. crassiceps infection.

           FIGURE 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 7.

Superoxide and H2O2 contribute to the antiproliferative capacity of adherent peritoneal cells from late stage T. crassiceps-infected mice. Adherent peritoneal cells from three infected and three noninfected mice were cocultured with lymph node cells from noninfected mice activated with Con A in the absence or presence of superoxide (SOD) or H2O2 (catalase) inhibitor. Proliferation was determined by [3H]Thy incorporation. The mean ± SD of infected and noninfected mice, of one representative of three independent experiments performed in the late stage of infection (> 6 wk), is shown. ∗, p < 0.05 lower comparing infected vs noninfected mice. #, p < 0.05 higher comparing infected mice in the presence vs the absence of inhibitor.

Together, these data suggest that in the early stage of T. crassiceps infection, classical MSC are responsible for the inhibition of T cell proliferation mainly via the secretion of NO. Yet, in the late stage of infection, alternative MSC block the proliferation in an arginase and iNOS-dependent pathway. In the whole course of the disease, the suppressive activity was reversed when the suppressive cell population was separated from responder T cells in transwell culture chambers. In addition, paraformaldehyde-fixed cells cocultured with responder cells fully retained their suppressive activity (data not shown). Therefore, as in B. malayi-infected mice (25), the suppressive activity in T. crassiceps-infected animals seems to be mediated by a direct cell contact. However, we cannot exclude that small molecules including NO and ROS still diffused through the membrane of fixed suppressive cells.

IL-4 and IL-13 trigger the expansion of alternative MSC

To address whether the induction of suppressive cells in the late stage of T. crassiceps infection depends on IL-4 and/or IL-13, experiments were performed in IL-4−/− and IL-R4α−/− mice. Parasite burdens in peritoneal lavages were similar in infected wild-type, IL-4−/−, and IL-R4α−/− mice at the time when cells were isolated (data not shown). In agreement with previous reports (25, 35), no suppressive activity of adherent peritoneal cells on T cell proliferation triggered by Con A was recorded in the absence of IL-4 (IL-4−/− mice) or of IL-4 and IL-13 (IL-4Rα−/− mice) (data not shown). Correspondingly, real-time PCR analyses revealed that arginase mRNA expression in adherent peritoneal cells was not induced upon infection in IL-4−/− and IL-4Rα−/− mice, in contrast to wild-type animals (Table III⇓). In addition, the induction of FIZZ1 and MGL expression was abolished in the absence of IL-4 and/or IL-13 signaling in infected mice. Finally, the induction of Ym expression observed in infected wild-type animals was lowered drastically in IL-4−/− and even more in IL-4Rα−/−-infected mice.

View this table:
  • View inline
  • View popup
Table III.

Modulation of arginase, FIZZ1, Ym, MGL, and 12/15-LOX mRNA expression in adherent peritoneal cells of wild-type, IL-4−/−, and IL-4Rα−/− mice in the late stage of T. crassiceps infectiona

The contribution of IL-4 and IL-13 to the accumulation of CD11b+GR-1+ cells in T. crassiceps-infected mice was investigated in the late stage of infection (Fig. 8⇓ and Table IV⇓). In noninfected wild-type, IL-4−/− and IL-4Rα−/− mice, the percentages of CD11b+ populations, including GR-1−, GR-1low, and GR-1high cells, were similar. In infected animals, the percentages of CD11b+ cells, as compared with wild-type mice, slightly decreased in IL-4−/− mice and were significantly lower in IL-4Rα−/− mice. Notably, the percentages of the GR-1− cells expressing high or low levels of CD11b, and of CD11blowGR-1low cells, were significantly lower in infected IL-4−/− and IL-4Rα−/− mice than in infected wild-type mice. In contrast, the percentage of CD11blowGR-1high cells increased both in infected IL-4−/− and IL-4Rα−/− mice. Finally, a population of CD11b−GR-1low cells, which was marginally present in the three strains of noninfected mice and in infected wild-type mice, expanded in IL-4−/− and IL-4Rα−/− mice. The decrease in CD11bhighGR-1−, CD11blowGR-1−, and CD11blowGR-1low cells concomitant with the increase in CD11blowGR-1high and CD11b−GR-1low cells already occurred in the early stage of infection (data not shown).

           FIGURE 8.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 8.

IL-4 and/or IL-13 contribute to the expansion of CD11b+GR-1+ cells in the peritoneal cavity of late stage T. crassiceps-infected mice. In the late stage of infection (>6 wk), cells from the peritoneal cavity were collected in individual wild-type, IL-4−/−, and IL-4Rα−/− mice and stained with FITC-labeled anti-CD11b and PE-labeled anti-GR-1 Abs. Simultaneously, cells from three noninfected mice were pooled and submitted to the same staining procedure. The percentage of gated positive cells within the total cell population was determined. Profiles are representative of one pool of cells from noninfected mice and of cells from one infected individual of six investigated in two independent experiments.

View this table:
  • View inline
  • View popup
Table IV.

Percentage of CD11b+ and GR-1+ cells in the peritoneal cavity of wild-type, IL-4−/− and IL-4Rα−/− mice in the late stage of T. crassiceps infectiona

In accordance with a previous report (35), the populations expressing GR-1 that expanded in the absence of IL-4 and or IL-13 signaling in Taenia-infected mice should not be neutrophils. Indeed, the percentage of peritoneal myeloperoxidase-positive cells (cytochemical localization) and the gene expression levels of myeloperoxidase in adherent peritoneal cells (real-time PCR) were similar in knockout (KO) and wild-type mice both in the early and the late stage of infection (data not shown). Although not firmly established, these expanding GR-1+ populations may represent eosinophils (33) and/or B cells (34).

12/15-LOX activity contributes to the suppressive activity of alternative myeloid cells

IL-4 has been reported to interfere with T cell proliferation by inducing the macrophage 12/15-LOX activity, hereby generating potential ligands for peroxisome proliferator-activated receptor-γ (PPAR-γ), a member of the ligand-dependent nuclear receptor family (43). To assess this possible pathway of suppression, 12/15-LOX gene expression was evaluated by real-time PCR in adherent peritoneal cells collected in the late stage of T. crassiceps infection. As shown in Table III⇑, 12/15-LOX gene expression was similar in infected and noninfected wild-type animals. However, in infected IL-4−/− and IL-4Rα−/− mice, the expression of 12/15-LOX mRNA was abolished completely.

Next, the suppressive activity of adherent peritoneal cells from late stage infection was investigated in the presence of NDGA, a general LOX inhibitor, or of GW9662, as a PPAR-γ antagonist. As shown in Fig. 9⇓, the Con A-induced T cell proliferation was restored by the LOX or the PPAR-γ inhibitor (by 55 ± 9% or 62 ± 11%, respectively). Although ligands for PPAR-γ can derive from the cyclooxygenase (COX) cascade (44), the nonselective COX inhibitor indomethacin did not influence the suppressive activity of adherent peritoneal cells from T. crassiceps-infected mice (data not shown).

           FIGURE 9.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 9.

The 12/15-LOX and PPAR-γ activation contribute to the antiproliferative capacity of adherent peritoneal cells from late stage T. crassiceps-infected mice. Adherent peritoneal cells from three infected and three noninfected mice were cocultured with lymph node cells from noninfected mice activated with Con A in the absence or presence of NDGA (LOX inhibitor) or GW9662 (PPAR-γ inhibitor). Proliferation was determined by [3H]Thy incorporation. The mean ± SD of infected and noninfected mice of one representative of three independent experiments performed in the late stage of infection (>6 wk) is shown. ∗, p < 0.05 lower comparing infected vs noninfected mice. #, p < 0.05 higher comparing infected mice in the presence vs the absence of inhibitor.

Together, these data indicate that IL-4/IL-13 contribute to the suppressive activity elicited during T. crassiceps infection through 12/15-LOX activation, resulting in the production of PPAR-γ ligands. This mechanism of suppression, as those induced by arginase and ROS, are only active in the late stage of infection (data not shown), i.e., when only alternative MSC are elicited.

It is worthwhile mentioning that, as with Con A, the inhibition of anti-CD3 Ab-induced T cell proliferation depended on iNOS for MSC elicited in the early stage of infection. In contrast, in the late stage of infection, arginase plus iNOS, superoxide, H2O2, LOX, and PPAR-γ contributed to the impairment of TCR-triggered T cell proliferation caused by alternative MSC (data not shown).

Discussion

The helminth Taenia solium causes neurocysticercosis in humans and pigs. This disease is a major public health problem in South America and Asia, and the number of reported cases in developed countries is increasing currently (45). In experimental models of cysticercosis, T. crassiceps-infected mice gradually shift from an early Th1/Th2 immune response to a Th2-biased immune response in the late stage of the disease. As one of the exceptions to the canonical believe that a Th2 immune response protects against helminth infections, a Th1 immune response is essential for host defense against T. crassiceps. In this parasitic disease, the Th2-biased immune response is associated with an increase in parasite load (27, 35, 46).

Recent studies suggest that a subset of macrophages called alternatively activated macrophages play a role in the bias toward the Th2 immune response induced by T. crassiceps or other helminths. Concomitantly, they weaken the host immune status by suppressing helminth-related and -unrelated immune responses. Such immunosuppression was proposed to impair the induction of the Th1 immune response, to increase the susceptibility of both mice and human to other diseases, or to limit the efficacy of vaccination in infected populations (25, 27, 47, 48, 49, 50, 51). In this study, we have further investigated the phenotype and mechanisms by which cells from the peritoneal cavity of mice implanted with T. crassiceps down-regulate T cell proliferation.

In accordance with previous microscopic analyses (26, 27, 35), we observed that as T. crassiceps infection progresses, a gradual decrease in the percentage of T and B cells and an increase in the percentage of CD11blowGR-1high cells representing granulocytes occurs. We also document that the percentage of F4/80highCD11bhigh mature macrophages tends to decrease. Concomitantly, a population of CD11blowGR-1low immature myeloid cells, which is marginally present in the peritoneal cavity of noninfected mice, expands gradually in the course of Taenia infection.

Adherent peritoneal cells from T. crassiceps-infected mice inhibit the proliferative response of T cells from noninfected mice triggered by Con A in a cell contact-dependent manner. The suppressive activity in this population, exhibiting in the course of infection an increase in CD11blowF4/80low cells expressing different levels of GR-1 (high or low), could thus be due to granulocytes, immature myeloid cells, or both. We believe that CD11blowGR-1low cells are the most likely suppressive cells. Indeed, their surface phenotype corresponds to the one of MSC, the suppressive activity of which was established clearly in various pathologies (20). Moreover, we document that the disappearance of suppressive activity in IL-4−/− and IL-4Rα−/− mice infected with T. crassiceps correlates with the loss of CD11blowGR-1low cells and the expansion of CD11blowGR-1high granulocytes.

The activation of peritoneal MSC in T. crassiceps-infected mice changes from a classical/alternative status in the early stage of infection to a predominant alternative status in the late stage of infection. The apoptosis of T and B cells observed in the early stage of T. crassiceps infection (data not shown and Ref. 52) as well as parasite-derived glycoconjugate and/or protein moieties may contribute to the emergence of alternative MSC, as described in other infection models (10, 53, 54, 55, 56, 57, 58). Reflecting the change in the activation status, MSC from the early or late stage of infection trigger T cells to secrete Th1/Th2 or Th2 cytokines, respectively. In addition, mechanisms underlying their antiproliferative function modify in the course of infection. Early stage MSC, despite exhibiting iNOS and arginase activity, block T cell proliferation only through their secretion of NO. This is evocative of classical MSC induced in tumor- or Trypanosoma cruzi-induced suppression (13, 15, 19, 59). In the late stage of infection, alternative MSC from T. crassiceps-infected animals impair T cell proliferation via their arginase activity. Yet, the combination of arginase and NO inhibitors was required to fully restore the T cell proliferative response. Moreover, alternative MSC impair T cell proliferative response by producing superoxide and H2O2. These mechanisms are reminiscent of alternative MSC elicited in cancer models (22, 23, 32). The current working mechanism is that arginase activity, by depleting l-arginine in alternative MSC, forces the iNOS reductase activity. This ends in the production of ROS, including superoxide and H2O2 that impair T cell proliferation. It should be remarked that the iNOS inhibitor l-NMMA could contribute to the depletion of arginine in alternative MSC by competing with CAT-2B, the transporter of l-arginine within cells (60).

IL-4 and IL-13 support the expansion of early myeloid progenitors and are required for the recruitment of immune cells in lymphoid tissues (61, 62, 63). Fittingly, the present and other reports demonstrate that IL-4 and/or IL-13 signaling is required for the generation of suppressive cells in the peritoneal cavity of helminth-infected mice (25, 26, 29, 35, 38, 64). Our data show that both cytokines could contribute to the antiproliferative capacity of T. crassiceps-induced alternative MSC by triggering arginase gene expression. Moreover, IL-4/IL-13 may play a role in the suppressive activity of alternative MSC by allowing the expression of the 12/15-LOX gene. The 12/15-LOX, previously associated with alternative macrophage activation (65), generates bioactive lipid mediators like 13-hydroxyoctadecadienoic acid and 15-hydroxyeicosatetraenoic acid, which serve as ligands for PPAR-γ. This nuclear hormone receptor, initially identified as a key mediator of lipid metabolism, exerts immunoregulatory functions at the level of both macrophages and T cells (66). When activated in T lymphocytes, PPAR-γ inhibits their proliferation by promoting the inhibition of IL-2 production (43, 67, 68). Accordingly, in T. crassiceps-infected animals, the inhibition of proliferation to Con A was shown to result from impaired IL-2 production (40, 42). In this article, we show that the suppressive capacity of alternative MSC is impaired partially by a LOX and PPAR-γ inhibitor. These observations support the idea that in T. crassiceps infection, IL-4/IL-13-activated alternative MSC exert 12/15-LOX activity, hereby secreting molecules that inhibit proliferation via PPAR-γ activation in T cells. In contrast, a COX inhibitor does not affect the suppressive potential of cells from T. crassiceps-infected mice. Thus, ligands of PPAR-γ like 15-deoxy-Δ12,14 prostaglandin. J2 that can be produced via the COX pathway (44) do not play a role in the antiproliferative activity of alternative MSC. Besides a role in the generation of PPAR-γ ligands, 12/15-LOX could also contribute to the production of ROS (69).

In summary, our study reveals the expansion of CD11blowGR-1low MSC in the course of T. crassiceps infection in mice. Early stage MSC, displaying the characteristics of classical and alternative activation, impair T cell proliferation via NO secretion, whereas the suppressive activity of alternative MSC elicited in the late stage of infection depends on the production of ROS via arginase activity and the production of PPAR-γ ligands via 12/15-LOX activity. Finally, IL-4/IL-13 signaling contributes to the expansion and suppressive activity of MSC. Therefore, this work proposes new intervention strategies to reverse immunosuppression, hereby restoring the efficacy of vaccination and resistance to opportunistic diseases in helminth-infected hosts.

Disclosures

The authors have no financial conflict of interest.

Footnotes

  • The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

  • ↵1 This work was supported by grants from the Fund for Scientific Research Flanders, the Institute for Promotion of Innovation by Science and Technology in Flanders (Generisch Basisonderzoek aan de Universiteiten), the United Nations Development Program/World Bank/World Health Organization Special Program for Research and Training in Tropical Diseases (Tropical Disease Research), and a Bilateral International Scientific and Technological Cooperation Grant from the Ministry of the Flemish Community, and was performed in frame of an Interuniversity Attraction Pole Program. F.B. received a Wellcome Trust Senior Fellowship for Medical Science in South Africa (Grant No. 056708/Z/99).

  • ↵2 Address correspondence and reprint requests to Dr. Alain Beschin, Department of Cellular and Molecular Interactions, Vlaams Interuniversitair Instituut voor Biotechnologie, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium. E-mail address: abeschin{at}vub.ac.be

  • ↵3 Abbreviations used in this paper: MSC, myeloid suppressor cell; iNOS, inducible NO synthase; ROS, reactive oxygen species; 12/15-LOX, 12/15-lipoxygenase; FIZZ1, found in inflammatory zone 1; l-NMMA, NG-monomethyl l-arginine; nor-NOHA, N(omega)-hydroxy-nor-l-arginine; SOD, superoxide dismutase; NDGA, nordihydroguaiaretic acid; [3H]Thy, [methyl-3H]-thymidine; MGL, macrophage galactose-type C-type lectin; KO, knockout; PPAR-γ, peroxisome proliferator-activated receptor-γ; COX, cyclooxygenase.

  • Received September 15, 2004.
  • Accepted February 27, 2005.
  • Copyright © 2005 by The American Association of Immunologists

References

  1. ↵
    Strober, S.. 1984. Natural suppressor (NS) cells, neonatal tolerance, and total lymphoid irradiation: exploring obscure relationships. Annu. Rev. Immunol. 2: 219-237.
    OpenUrlCrossRefPubMed
  2. ↵
    Maier, T., J. H. Holda, H. N. Claman. 1989. Natural suppressor cells. Prog. Clin. Biol. Res. 288: 235-244.
    OpenUrlPubMed
  3. ↵
    Pelaez, B., J. A. Campillo, J. A. Lopez-Asenjo, J. L. Subiza. 2001. Cyclophosphamide induces the development of early myeloid cells suppressing tumor cell growth by a nitric oxide-dependent mechanism. J. Immunol. 166: 6608-6615.
    OpenUrlAbstract/FREE Full Text
  4. ↵
    Serafini, P., C. De Santo, I. Marigo, S. Cingarlini, L. Dolcetti, G. Gallina, P. Zanovello, V. Bronte. 2004. Derangement of immune responses by myeloid suppressor cells. Cancer Immunol. Immunother. 53: 64-72.
    OpenUrlCrossRefPubMed
  5. ↵
    Ferret-Bernard, S., P. Sai, J. M. Bach. 2004. In vitro induction of inhibitory macrophage differentiation by granulocyte-macrophage colony-stimulating factor, stem cell factor and interferon-γ from lineage phenotypes-negative c-kit-positive murine hematopoietic progenitor cells. Immunol. Lett. 91: 221-217.
    OpenUrlCrossRefPubMed
  6. ↵
    Bronte, V., E. Apolloni, A. Cabrelle, R. Ronca, P. Serafini, P. Zamboni, N. P. Restifo, P. Zanovello. 2000. Identification of a CD11b+/Gr-1+/CD31+ myeloid progenitor capable of activating or suppressing CD8+ T cells. Blood 96: 3838-3846.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    Bronte, V., M. Wang, W. W. Overwijk, D. R. Surman, F. Pericle, S. A. Rosenberg, N. P. Restifo. 1998. Apoptotic death of CD8+ T lymphocytes after immunization: induction of a suppressive population of Mac-1+/Gr-1+ cells. J. Immunol. 161: 5313-5320.
    OpenUrlAbstract/FREE Full Text
  8. ↵
    Atochina, O., T. Daly-Engel, D. Piskorska, E. McGuire, D. A. Harn. 2001. A schistosome-expressed immunomodulatory glycoconjugate expands peritoneal Gr1+ macrophages that suppress naive CD4+ T cell proliferation via an IFN-γ and nitric oxide-dependent mechanism. J. Immunol. 167: 4293-4302.
    OpenUrlAbstract/FREE Full Text
  9. ↵
    Cauley, L. S., E. E. Miller, M. Yen, S. L. Swain. 2000. Superantigen-induced CD4 T cell tolerance mediated by myeloid cells and IFN-γ. J. Immunol. 165: 6056-6066.
    OpenUrlAbstract/FREE Full Text
  10. ↵
    Terrazas, L. I., K. L. Walsh, D. Piskorska, E. McGuire, D. A. Harn, Jr. 2001. The schistosome oligosaccharide lacto-N-neotetraose expands Gr1+ cells that secrete anti-inflammatory cytokines and inhibit proliferation of naive CD4+ cells: a potential mechanism for immune polarization in helminth infections. J. Immunol. 167: 5294-5303.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    Angulo, I., F. G. de las Heras, J. F. Garcia-Bustos, D. Gargallo, M. A. Munoz-Fernandez, M. Fresno. 2000. Nitric oxide-producing CD11b+Ly-6G(Gr-1)+CD31(ER-MP12)+ cells in the spleen of cyclophosphamide-treated mice: implications for T-cell responses in immunosuppressed mice. Blood 95: 212-220.
    OpenUrlAbstract/FREE Full Text
  12. ↵
    Bronte, V., D. B. Chappell, E. Apolloni, A. Cabrelle, M. Wang, P. Hwu, N. P. Restifo. 1999. Unopposed production of granulocyte-macrophage colony-stimulating factor by tumors inhibits CD8+ T cell responses by dysregulating antigen-presenting cell maturation. J. Immunol. 162: 5728-5737.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    Apolloni, E., V. Bronte, A. Mazzoni, P. Serafini, A. Cabrelle, D. M. Segal, H. A. Young, P. Zanovello. 2000. Immortalized myeloid suppressor cells trigger apoptosis in antigen-activated T lymphocytes. J. Immunol. 165: 6723-6730.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    Liu, Y., J. A. Van Ginderachter, L. Brys, P. De Baetselier, G. Raes, A. B. Geldhof. 2003. Nitric oxide-independent CTL suppression during tumor progression: association with arginase-producing (M2) myeloid cells. J. Immunol. 170: 5064-5074.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    Gabrilovich, D. I., M. P. Velders, E. M. Sotomayor, W. M. Kast. 2001. Mechanism of immune dysfunction in cancer mediated by immature Gr-1+ myeloid cells. J. Immunol. 166: 5398-5406.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    Li, Q., P. Y. Pan, P. Gu, D. Xu, S. H. Chen. 2004. Role of immature myeloid Gr-1+ cells in the development of antitumor immunity. Cancer Res. 64: 1130-1139.
    OpenUrlAbstract/FREE Full Text
  17. ↵
    Mencacci, A., C. Montagnoli, A. Bacci, E. Cenci, L. Pitzurra, A. Spreca, M. Kopf, A. H. Sharpe, L. Romani. 2002. CD80+Gr-1+ myeloid cells inhibit development of antifungal Th1 immunity in mice with candidiasis. J. Immunol. 169: 3180-3190.
    OpenUrlAbstract/FREE Full Text
  18. ↵
    Mordue, D. G., L. D. Sibley. 2003. A novel population of Gr-1+-activated macrophages induced during acute toxoplasmosis. J. Leukocyte Biol. 74: 1015-1025.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Goni, O., P. Alcaide, M. Fresno. 2002. Immunosuppression during acute Trypanosoma cruzi infection: involvement of Ly6G (Gr1+)CD11b+immature myeloid suppressor cells. Int. Immunol. 14: 1125-1134.
    OpenUrlAbstract/FREE Full Text
  20. ↵
    Bronte, V., P. Serafini, A. Mazzoni, D. M. Segal, P. Zanovello. 2003. l-arginine metabolism in myeloid cells controls T-lymphocyte functions. Trends Immunol. 24: 302-306.
    OpenUrlCrossRefPubMed
  21. ↵
    Terabe, M., J. A. Berzofsky. 2004. Immunoregulatory T cells in tumor immunity. Curr. Opin. Immunol. 16: 157-162.
    OpenUrlCrossRefPubMed
  22. ↵
    Terabe, M., S. Matsui, J. M. Park, M. Mamura, N. Noben-Trauth, D. D. Donaldson, W. Chen, S. M. Wahl, S. Ledbetter, B. Pratt, et al 2003. Transforming growth factor-β production and myeloid cells are an effector mechanism through which CD1d-restricted T cells block cytotoxic T lymphocyte-mediated tumor immunosurveillance: abrogation prevents tumor recurrence. J. Exp. Med. 198: 1741-1752.
    OpenUrlAbstract/FREE Full Text
  23. ↵
    Kusmartsev, S., Y. Nefedova, D. Yoder, D. I. Gabrilovich. 2004. Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J. Immunol. 172: 989-999.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    Loke, P., A. S. MacDonald, J. E. Allen. 2000. Antigen-presenting cells recruited by Brugia malayi induce Th2 differentiation of naive CD4+ T cells. Eur. J. Immunol. 30: 1127-1135.
    OpenUrlCrossRefPubMed
  25. ↵
    Loke, P., A. S. MacDonald, A. Robb, R. M. Maizels, J. E. Allen. 2000. Alternatively activated macrophages induced by nematode infection inhibit proliferation via cell-to-cell contact. Eur. J. Immunol. 30: 2669-2678.
    OpenUrlCrossRefPubMed
  26. ↵
    MacDonald, A. S., R. M. Maizels, R. A. Lawrence, I. Dransfield, J. E. Allen. 1998. Requirement for in vivo production of IL-4, but not IL-10, in the induction of proliferative suppression by filarial parasites. J. Immunol. 160: 1304-1312.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Rodriguez-Sosa, M., A. R. Satoskar, R. Calderon, L. Gomez-Garcia, R. Saavedra, R. Bojalil, L. I. Terrazas. 2002. Chronic helminth infection induces alternatively activated macrophages expressing high levels of CCR5 with low interleukin-12 production and Th2-biasing ability. Infect. Immun. 70: 3656-3664.
    OpenUrlAbstract/FREE Full Text
  28. ↵
    Rodriguez-Sosa, M., L. E. Rosas, J. R. David, R. Bojalil, A. R. Satoskar, L. I. Terrazas. 2003. Macrophage migration inhibitory factor plays a critical role in mediating protection against the helminth parasite Taenia crassiceps. Infect. Immun. 71: 1247-1254.
    OpenUrlAbstract/FREE Full Text
  29. ↵
    Marshall, M. A., D. Jankovic, V. E. Maher, A. Sher, J. A. Berzofsky. 2001. Mice infected with Schistosoma mansoni develop a novel non-T-lymphocyte suppressor population which inhibits virus-specific CTL induction via a soluble factor. Microbes Infect. 3: 1051-1061.
    OpenUrlCrossRefPubMed
  30. ↵
    Noben-Trauth, N., G. Kohler, K. Burki, B. Ledermann. 1996. Efficient targeting of the IL-4 gene in a BALB/c embryonic stem cell line. Transgenic Res. 5: 487-491.
    OpenUrlCrossRefPubMed
  31. ↵
    Mohrs, M., B. Ledermann, G. Kohler, A. Dorfmuller, A. Gessner, F. Brombacher. 1999. Differences between IL-4- and IL-4 receptor α-deficient mice in chronic leishmaniasis reveal a protective role for IL-13 receptor signaling. J. Immunol. 162: 7302-7308.
    OpenUrlAbstract/FREE Full Text
  32. ↵
    Bronte, V., P. Serafini, C. De Santo, I. Marigo, V. Tosello, A. Mazzoni, D. M. Segal, C. Staib, M. Lowel, G. Sutter, et al 2003. IL-4-induced arginase 1 suppresses alloreactive T cells in tumor-bearing mice. J. Immunol. 170: 270-278.
    OpenUrlAbstract/FREE Full Text
  33. ↵
    Taylor, P. R., G. D. Brown, A. B. Geldhof, L. Martinez-Pomares, S. Gordon. 2003. Pattern recognition receptors and differentiation antigens define murine myeloid cell heterogeneity ex vivo. Eur. J. Immunol. 33: 2090-2097.
    OpenUrlCrossRefPubMed
  34. ↵
    Ramalingam, T., B. Rajan, J. Lee, T. V. Rajan. 2003. Kinetics of cellular responses to intraperitoneal Brugia pahangi infections in normal and immunodeficient mice. Infect. Immun. 71: 4361-4367.
    OpenUrlAbstract/FREE Full Text
  35. ↵
    Rodriguez-Sosa, M., J. R. David, R. Bojalil, A. R. Satoskar, L. I. Terrazas. 2002. Cutting edge: susceptibility to the larval stage of the helminth parasite Taenia crassiceps is mediated by Th2 response induced via STAT6 signaling. J. Immunol. 168: 3135-3139.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    Toenjes, S. A., R. E. Kuhn. 2003. The initial immune response during experimental cysticercosis is of the mixed Th1/Th2 type. Parasitol Res. 89: 407-413.
    OpenUrlCrossRefPubMed
  37. ↵
    Raes, G., L. Brys, B. K. Dahal, J. Brandt, J. Grooten, F. Brombacher, G. Vanham, W. Noel, P. Bogaert, T. Boonefaes, et al 2005. Macrophage galactose-type C-type lectins as novel markers for alternatively activated macrophages elicited by parasitic infections and allergic airway inflammation. J. Leukocyte Biol. 77: 321-327.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    Loke, P., M. G. Nair, J. Parkinson, D. Guiliano, M. Blaxter, J. E. Allen. 2002. IL-4 dependent alternatively-activated macrophages have a distinctive in vivo gene expression phenotype. BMC Immunol. 3: 7.
    OpenUrlCrossRefPubMed
  39. ↵
    Raes, G., P. De Baetselier, W. Noel, A. Beschin, F. Brombacher, G. Hassanzadeh Gh. 2002. Differential expression of FIZZ1 and Ym1 in alternatively versus classically activated macrophages. J. Leukocyte Biol. 71: 597-602.
    OpenUrlAbstract/FREE Full Text
  40. ↵
    Sciutto, E., G. Fragoso, M. Baca, V. De la Cruz, L. Lemus, E. Lamoyi. 1995. Depressed T-cell proliferation associated with susceptibility to experimental Taenia crassiceps infection. Infect. Immun. 63: 2277-2281.
    OpenUrlAbstract/FREE Full Text
  41. ↵
    Spolski, R. J., M. A. Alexander-Miller, R. E. Kuhn. 2002. Suppressed cytotoxic T lymphocyte responses in experimental cysticercosis. Vet. Parasitol. 106: 325-330.
    OpenUrlCrossRefPubMed
  42. ↵
    Spolski, R. J., J. Corson, P. G. Thomas, R. E. Kuhn. 2000. Parasite-secreted products regulate the host response to larval Taenia crassiceps. Parasite Immunol. 22: 297-305.
    OpenUrlCrossRefPubMed
  43. ↵
    Yang, X. Y., L. H. Wang, K. Mihalic, W. Xiao, T. Chen, P. Li, L. M. Wahl, W. L. Farrar. 2002. Interleukin (IL)-4 indirectly suppresses IL-2 production by human T lymphocytes via peroxisome proliferator-activated receptor γ activated by macrophage-derived 12/15-lipoxygenase ligands. J. Biol. Chem. 277: 3973-3978.
    OpenUrlAbstract/FREE Full Text
  44. ↵
    Na, H. K., Y. J. Surh. 2003. Peroxisome proliferator-activated receptor γ (PPARγ) ligands as bifunctional regulators of cell proliferation. Biochem. Pharmacol. 66: 1381-1391.
    OpenUrlCrossRefPubMed
  45. ↵
    Sciutto, E., G. Fragoso, A. Fleury, J. P. Laclette, J. Sotelo, A. Aluja, L. Vargas, C. Larralde. 2000. Taenia solium disease in humans and pigs: an ancient parasitosis disease rooted in developing countries and emerging as a major health problem of global dimensions. Microbes Infect. 2: 1875-1890.
    OpenUrlCrossRefPubMed
  46. ↵
    Terrazas, L. I., M. Cruz, M. Rodriguez-Sosa, R. Bojalil, F. Garcia-Tamayo, C. Larralde. 1999. Th1-type cytokines improve resistance to murine cysticercosis caused by Taenia crassiceps. Parasitol. Res. 85: 135-141.
    OpenUrlCrossRefPubMed
  47. ↵
    Hesse, M., M. Modolell, A. C. La Flamme, M. Schito, J. M. Fuentes, A. W. Cheever, E. J. Pearce, T. A. Wynn. 2001. Differential regulation of nitric oxide synthase-2 and arginase-1 by type 1/type 2 cytokines in vivo: granulomatous pathology is shaped by the pattern of l-arginine metabolism. J. Immunol. 167: 6533-6544.
    OpenUrlAbstract/FREE Full Text
  48. ↵
    Kullberg, M. C., E. J. Pearce, S. E. Hieny, A. Sher, J. A. Berzofsky. 1992. Infection with Schistosoma mansoni alters Th1/Th2 cytokine responses to a non-parasite antigen. J. Immunol. 148: 3264-3270.
    OpenUrlAbstract
  49. ↵
    Actor, J. K., M. A. Marshall, I. A. Eltoum, R. M. Buller, J. A. Berzofsky, A. Sher. 1994. Increased susceptibility of mice infected with Schistosoma mansoni to recombinant vaccinia virus: association of viral persistence with egg granuloma formation. Eur. J. Immunol. 24: 3050-3056.
    OpenUrlCrossRefPubMed
  50. ↵
    Kamal, S., M. Madwar, L. Bianchi, A. E. Tawil, R. Fawzy, T. Peters, J. W. Rasenack. 2000. Clinical, virological and histopathological features: long-term follow-up in patients with chronic hepatitis C co-infected with S. mansoni. Liver 20: 281-289.
    OpenUrlCrossRefPubMed
  51. ↵
    Sabin, E. A., M. I. Araujo, E. M. Carvalho, E. J. Pearce. 1996. Impairment of tetanus toxoid-specific Th1-like immune responses in humans infected with Schistosoma mansoni. J. Infect. Dis. 173: 269-272.
    OpenUrlAbstract/FREE Full Text
  52. ↵
    Lopez-Briones, S., E. Sciutto, J. L. Ventura, A. Zentella, G. Fragoso. 2003. CD4+ and CD19+ splenocytes undergo apoptosis during an experimental murine infection with Taenia crassiceps. Parasitol. Res. 90: 157-163.
    OpenUrlPubMed
  53. ↵
    Freire-de-Lima, C. G., D. O. Nascimento, M. B. Soares, P. T. Bozza, H. C. Castro-Faria-Neto, F. G. de Mello, G. A. DosReis, M. F. Lopes. 2000. Uptake of apoptotic cells drives the growth of a pathogenic trypanosome in macrophages. Nature 403: 199-203.
    OpenUrlCrossRefPubMed
  54. ↵
    Ribeiro-Gomes, F. L., A. C. Otero, N. A. Gomes, M. C. Moniz-De-Souza, L. Cysne-Finkelstein, A. C. Arnholdt, V. L. Calich, S. G. Coutinho, M. F. Lopes, G. A. DosReis. 2004. Macrophage interactions with neutrophils regulate Leishmania major infection. J. Immunol. 172: 4454-4462.
    OpenUrlAbstract/FREE Full Text
  55. ↵
    Tawill, S., L. Le Goff, F. Ali, M. Blaxter, J. E. Allen. 2004. Both free-living and parasitic nematodes induce a characteristic Th2 response that is dependent on the presence of intact glycans. Infect. Immun. 72: 398-407.
    OpenUrlAbstract/FREE Full Text
  56. ↵
    Stempin, C., L. Giordanengo, S. Gea, F. Cerban. 2002. Alternative activation and increase of Trypanosoma cruzi survival in murine macrophages stimulated by cruzipain, a parasite antigen. J. Leukocyte Biol. 72: 727-734.
    OpenUrlAbstract/FREE Full Text
  57. ↵
    Giordanengo, L., N. Guinazu, C. Stempin, R. Fretes, F. Cerban, S. Gea. 2002. Cruzipain, a major Trypanosoma cruzi antigen, conditions the host immune response in favor of parasite. Eur. J. Immunol. 32: 1003-1011.
    OpenUrlCrossRefPubMed
  58. ↵
    Stempin, C. C., T. B. Tanos, O. A. Coso, F. M. Cerban. 2004. Arginase induction promotes Trypanosoma cruzi intracellular replication in Cruzipain-treated J774 cells through the activation of multiple signaling pathways. Eur. J. Immunol. 34: 200-209.
    OpenUrlCrossRefPubMed
  59. ↵
    Mazzoni, A., V. Bronte, A. Visintin, J. H. Spitzer, E. Apolloni, P. Serafini, P. Zanovello, D. M. Segal. 2002. Myeloid suppressor lines inhibit T cell responses by an NO-dependent mechanism. J. Immunol. 168: 689-695.
    OpenUrlAbstract/FREE Full Text
  60. ↵
    Closs, E. I., F. Z. Basha, A. Habermeier, U. Forstermann. 1997. Interference of l-arginine analogues with l-arginine transport mediated by the y+ carrier hCAT-2B. Nitric Oxide 1: 65-73.
    OpenUrlCrossRefPubMed
  61. ↵
    Jacobsen, S. E., C. Okkenhaug, O. P. Veiby, D. Caput, P. Ferrara, A. Minty. 1994. Interleukin 13: novel role in direct regulation of proliferation and differentiation of primitive hematopoietic progenitor cells. J. Exp. Med. 180: 75-82.
    OpenUrlAbstract/FREE Full Text
  62. ↵
    Snoeck, H. W., M. Lenjou, G. Nys, F. Lardon, M. E. Peetermans, D. R. Van Bockstaele, A. Moulijn, L. Haenen, Z. N. Berneman. 1996. Interleukin 4 and interferon γ costimulate the expansion of early human myeloid colony-forming cells. Proposal of a model for the regulation of myelopoiesis by interleukin 4 and interferon γ and its integration with the regulation of the immune response. Leukemia 10: 117-122.
    OpenUrlPubMed
  63. ↵
    Voehringer, D., K. Shinkai, R. M. Locksley. 2004. Type 2 immunity reflects orchestrated recruitment of cells committed to IL-4 production. Immunity 20: 267-277.
    OpenUrlCrossRefPubMed
  64. ↵
    La Flamme, A. C., E. A. Patton, B. Bauman, E. J. Pearce. 2001. IL-4 plays a crucial role in regulating oxidative damage in the liver during schistosomiasis. J. Immunol. 166: 1903-1911.
    OpenUrlAbstract/FREE Full Text
  65. ↵
    Gordon, S.. 2003. Alternative activation of macrophages. Nat. Rev. Immunol. 3: 23-35.
    OpenUrlCrossRefPubMed
  66. ↵
    Nencioni, A., S. Wesselborg, P. Brossart. 2003. Role of peroxisome proliferator-activated receptor γ and its ligands in the control of immune responses. Crit. Rev. Immunol. 23: 1-13.
    OpenUrlCrossRefPubMed
  67. ↵
    Huang, J. T., J. S. Welch, M. Ricote, C. J. Binder, T. M. Willson, C. Kelly, J. L. Witztum, C. D. Funk, D. Conrad, C. K. Glass. 1999. Interleukin-4-dependent production of PPAR-γ ligands in macrophages by 12/15-lipoxygenase. Nature 400: 378-382.
    OpenUrlCrossRefPubMed
  68. ↵
    Yang, X. Y., L. H. Wang, T. Chen, D. R. Hodge, J. H. Resau, L. DaSilva, W. L. Farrar. 2000. Activation of human T lymphocytes is inhibited by peroxisome proliferator-activated receptor γ (PPARγ) agonists. PPARγ co-association with transcription factor NFAT. J. Biol. Chem. 275: 4541-4544.
    OpenUrlAbstract/FREE Full Text
  69. ↵
    Lovat, P. E., S. Oliverio, M. Ranalli, M. Corazzari, C. Rodolfo, F. Bernassola, K. Aughton, M. Maccarrone, Q. D. Hewson, A. D. Pearson, et al 2002. GADD153 and 12-lipoxygenase mediate fenretinide-induced apoptosis of neuroblastoma. Cancer Res. 62: 5158-5167.
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top

In this issue

The Journal of Immunology: 174 (10)
The Journal of Immunology
Vol. 174, Issue 10
15 May 2005
  • Table of Contents
  • About the Cover
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word about The Journal of Immunology.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Reactive Oxygen Species and 12/15-Lipoxygenase Contribute to the Antiproliferative Capacity of Alternatively Activated Myeloid Cells Elicited during Helminth Infection
(Your Name) has forwarded a page to you from The Journal of Immunology
(Your Name) thought you would like to see this page from the The Journal of Immunology web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Reactive Oxygen Species and 12/15-Lipoxygenase Contribute to the Antiproliferative Capacity of Alternatively Activated Myeloid Cells Elicited during Helminth Infection
Lea Brys, Alain Beschin, Geert Raes, Gholamreza Hassanzadeh Ghassabeh, Wim Noël, Jef Brandt, Frank Brombacher, Patrick De Baetselier
The Journal of Immunology May 15, 2005, 174 (10) 6095-6104; DOI: 10.4049/jimmunol.174.10.6095

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Reactive Oxygen Species and 12/15-Lipoxygenase Contribute to the Antiproliferative Capacity of Alternatively Activated Myeloid Cells Elicited during Helminth Infection
Lea Brys, Alain Beschin, Geert Raes, Gholamreza Hassanzadeh Ghassabeh, Wim Noël, Jef Brandt, Frank Brombacher, Patrick De Baetselier
The Journal of Immunology May 15, 2005, 174 (10) 6095-6104; DOI: 10.4049/jimmunol.174.10.6095
del.icio.us logo Digg logo Reddit logo Twitter logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Disclosures
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Innate Immunity Together with Duration of Antigen Persistence Regulate Effector T Cell Induction
  • Regulatory Roles of IL-2 and IL-4 in H4/Inducible Costimulator Expression on Activated CD4+ T Cells During Th Cell Development
  • Induction of CD4+ T Cell Apoptosis as a Consequence of Impaired Cytoskeletal Rearrangement in UVB-Irradiated Dendritic Cells
Show more CELLULAR IMMUNOLOGY AND IMMUNE REGULATION

Similar Articles

Navigate

  • Home
  • Current Issue
  • Next in The JI
  • Archive
  • Brief Reviews
  • Pillars of Immunology
  • Translating Immunology

For Authors

  • Submit a Manuscript
  • Instructions for Authors
  • About the Journal
  • Journal Policies
  • Editors

General Information

  • Advertisers
  • Subscribers
  • Rights and Permissions
  • Accessibility Statement
  • FAR 889
  • Privacy Policy
  • Disclaimer

Journal Services

  • Email Alerts
  • RSS Feeds
  • ImmunoCasts
  • Twitter

Copyright © 2022 by The American Association of Immunologists, Inc.

Print ISSN 0022-1767        Online ISSN 1550-6606