Role of the programmed Death-1 pathway in the suppressive activity of alternatively activated macrophages in experimental cysticercosis

https://doi.org/10.1016/j.ijpara.2005.06.003Get rights and content

Abstract

We characterised a population of macrophages potentially involved in the immunoregulation induced by experimental cysticercosis. Following Taenia crassiceps infection, macrophages recruited in the peritoneal cavity were isolated and co-cultured at different ratios with T cells from naïve mice previously stimulated with anti-CD3/CD28 antibodies; these macrophages inhibited naïve T cell proliferation. This suppressive effect was Interleukin (IL)-10, Interferon-gamma (IFN-γ), and nitric oxide (NO) independent. In contrast, macrophage-T cell contact was necessary to maintain anergy of T cells. Reverse transcriptase-PCR analysis of these macrophages showed higher transcripts of IL-10, chitinases Fizz1 and Ym1, and arginase-1 compared with naïve macrophages; by contrast, IL-12p40, and inducible nitric oxide synthase (iNOS) transcripts were undetected, whereas C-C chemokine ligand 5 (CCL5) was unchanged. Analysis of the membrane molecules expressed on Taenia-induced macrophages showed an up-regulation of several markers, mainly programmed death ligand 1 (PD-L1) and PD-L2. Blockade of PD-L1, PD-L2 or their receptor PD-1, but not of another marker, eliminated their ability to inhibit T-cell proliferation. Parallel experiments using ovalbumin (OVA)-peptide as a model antigen displayed similar results. Additionally, the same mechanism appears to be functional in splenocytes of T. crassiceps-infected mice given that blockade of PD-1, PD-L1 or PD-L2 re-established their ability to proliferate in response to parasite antigens. Moreover, Taenia-induced macrophages were able to suppress a mixed lymphocyte reaction in a PD-1-dependent manner. Thus, cestode infections induce macrophages alternatively activated with strong suppressive activity involving the PD-1/PD-L's pathway.

Introduction

Parasitic helminths have developed complex mechanisms to evade their host's responses (Gause et al., 2003). Helminths and their antigens are now recognised to have important immunomodulatory activities (Maizels and Yazdanbakhsh, 2003). Commonly, helminth infections induce Th2-biased immune responses in their hosts that have been associated with impaired proliferative cell responses to parasitic antigens or unrelated antigens (Holland et al., 2000, Gause et al., 2003). Moreover, helminth infections can alter the immunity and susceptibility to other pathogens (Cox, 2001) or reduce the effectiveness of vaccines, such as tetanus vaccination during lymphatic filariasis (Nookala et al., 2004). How helminth infections modulate the host immune system remains unclear.

With few exceptions (Spolski et al., 2000, Hernandez-Mendoza et al., 2005), research on mechanisms of immunomodulation during cestode infections has been largely neglected. We have recently examined the immunomodulatory activities of the cestode Taenia crassiceps. As in most helminth infections, experimental cysticercosis caused by T. crassiceps induces Th2-type biased immune responses (Villa and Kuhn, 1996, Terrazas et al., 1998, Toenjes et al., 1999) as well as anergy of splenocytes in response to both antigen-specific and polyclonal stimuli (Sciutto et al., 1995, Villa and Kuhn, 1996, Rodriguez-Sosa et al., 2003). Besides, increased susceptibility to Trypanosoma cruzi (Rodriguez et al., 1999) and to vaccinia virus (Spolski et al., 2002) was observed in mice previously infected with T. crassiceps. Thus, in addition to activating Th2-type biased responses, infection with T. crassiceps can regulate other lymphocyte functions in vivo, suggesting that this infection may induce strong modulation of the immune response.

Classical activation of T cells involves interaction between membranes of both antigen presenting cells (APC) and T cells (Sharpe and Freeman, 2002) where the interaction of major histocompatibility complex (MHC) and T-cell receptor (TCR), as well as co-stimulatory molecules such as B7-1 and B7-2 with CD28 activate T cell responses. On the other hand, inhibitory interactions can be induced if cytotoxic T lymphocyte antigen 4 (CTLA-4) on the surface of activated T cells binds to B7-1 or B7-2 on APC (Sharpe and Freeman, 2002). Thus, accessory molecules can stimulate or inhibit T cells. In the last few years, new members of the B7 family have been identified. Two of the most relevant have been programmed death ligand 1 (PD-L1) and PD-L2 (Khoury and Sayegh, 2004, Greenwald et al., 2005). These molecules bind to the receptor programmed death 1 (PD-1) which is expressed on activated T cells (B cells and myeloid cells also) and their interactions result in down-modulation of T cell responses (Latchman et al., 2004).

In an attempt to dissect out mechanisms that lead to immune alteration in experimental murine cysticercosis, we examined the role of macrophages induced in response to T. crassiceps infection in modulating T cell proliferation to different classes of stimuli such as, polyclonal activation by anti-CD3/CD28 mAb, antigen-specific stimulation to parasite or unrelated antigens and, CD4+ cell proliferative response in a mixed lymphocyte reaction (MLR). We found that Taenia-induced macrophages had the ability to suppress T cell proliferation in vitro in a cell contact-dependent manner via an increased expression of PD-L1 and PD-L2.

Section snippets

Mice

Six to eight-week-old female BALB/cAnN and C57BL/6 mice were purchased from Harlan Laboratories (México), and DO11.10 mice TCR transgenic mice on BALB/c background originally obtained from The Jackson Lab, were maintained in a pathogen free environment at FES-Iztacala, UNAM animal facility in accordance with Institutional and National guidelines.

Parasites and infection protocol

Metacestodes of T. crassiceps were harvested in sterile conditions from the peritoneal cavity of female BALB/c mice after 2–4 months of infection. The

Macrophages obtained from T. crassiceps-infected mice are suppressive

Fig. 1 shows that T cells in the absence of macrophages (data not shown), or in the presence of macrophages from naïve mice exhibited high levels of proliferation in response to anti-CD3/CD28 mAb stimulation. In contrast, addition of macrophages from T. crassiceps-infected mice, at both time points post-infection analysed (4 and 8 weeks), markedly suppressed proliferation of CD90+ cells, especially when higher ratios of macrophages were added into the cultures (Fig. 1). There was a significant

Discussion

Helminths are known to induce immune anergy, anti-inflammatory responses as well as Th2 biased responses (Gause et al., 2003, Maizels and Yazdanbakhsh, 2003). Different components present in helminths have been evaluated (Maizels et al., 2004). Excreted/secreted products as well as membrane molecules, proteins and carbohydrates have been shown to be responsible for some of these events (Okano et al., 1999, Holland et al., 2000, Deehan et al., 2002).

Previous studies suggest that macrophages can

Acknowledgements

This work was supported by CONACYT grant A-M 4158 and by DGAPA-PAPIIT-UNAM grant IN223003 and IN245004.

References (41)

  • L. Carter et al.

    PD-1:PD-L inhibitory pathway affects both CD4(+) and CD8(+) T cells and is overcome by IL-2

    Eur. J. Immunol.

    (2002)
  • F.E. Cox

    Concomitant infections, parasites and immune responses

    Parasitology

    (2001)
  • M.R. Deehan et al.

    Immunomodulatory properties of Ascaris suum glycosphingolipids–phosphorylcholine and non-phosphorylcholine-dependent effects

    Parasite Immunol.

    (2002)
  • S. Donnelly et al.

    Thioredoxin peroxidase secreted by Fasciola hepatica induces the alternative activation of macrophages

    Infect. Immun.

    (2005)
  • R.J. Greenwald et al.

    The B7 family revisited

    Annu. Rev. Immunol.

    (2005)
  • L. Hernandez-Mendoza et al.

    The implantation of Taenia solium metacestodes in mice induces down-modulation of T-cell proliferation and cytokine production

    Parasitol. Res.

    (2005)
  • M.J. Holland et al.

    Proteins secreted by the parasitic nematode Nippostrongylus brasiliensis act as adjuvants for Th2 responses

    Eur. J. Immunol.

    (2000)
  • S.L. James et al.

    Inducible nitric oxide synthase-deficient mice develop enhanced type 1 cytokine-associated cellular and humoral immune responses after vaccination with attenuated Schistosoma mansoni cercariae but display partially reduced resistance

    Infect. Immun.

    (1998)
  • Y. Latchman et al.

    PD-L2 is a second ligand for PD-1 and inhibits T cell activation

    Nat. Immunol.

    (2001)
  • Y.E. Latchman et al.

    PD-L1-deficient mice show that PD-L1 on T cells, antigen-presenting cells, and host tissues negatively regulates T cells

    PNAS

    (2004)
  • Cited by (114)

    • Mechanisms of gastrointestinal pathogenesis and landscape of intestinal immunity

      2022, Viral, Parasitic, Bacterial, and Fungal Infections: Antimicrobial, Host Defense, and Therapeutic Strategies
    View all citing articles on Scopus
    View full text