Elsevier

Molecular Immunology

Volume 55, Issues 3–4, October 2013, Pages 220-230
Molecular Immunology

Lymphocyte-derived microparticles induce bronchial epithelial cells’ pro-inflammatory cytokine production and apoptosis

https://doi.org/10.1016/j.molimm.2013.01.017Get rights and content

Abstract

Objective

The aim of this study was to determine if human CEM (human lymphoblastoma) T cell-derived microparticles (LMPs) could directly induce human bronchial epithelial cells (BECs) apoptosis and cytokine production. We also tested if LMPs phagocytosis by BECs played a role in mediating these effects.

Methods

We generated LMPs from CEM (human lymphoblastoma) T cells to investigate their effects on a human BEC cell line (16HBE) in vitro.

Results

BECs (16HBE cells) incubation with LMPs resulted in significant production of inflammation-associated cytokines, including tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6, and IL-8, in a dose- and time-dependent manner. LMPs also induced increased activities of caspase-3, caspase-8, and caspase-9 in BECS, which resulted in increased BECs apoptosis as assessed by flow cytometry (Annexin V and propidium iodide staining) and transmission electronic microscopy (TEM). Interestingly, LMPs effects on BECS were inhibited by the phagocytosis inhibitors cytochalasin D and chloroquine.

Conclusions

These results suggest that phagocytosis plays an important role in mediating the effects of LMPs on BECs. Thus, increased LMP concentrations may contribute to increased respiratory inflammatory responses and innate immune response maintenance in airway epithelium after LMPs engulfment by endothelial cells.

Highlights

► Incubation with LMPs increased production of inflammatory cytokines by BECs. ► LMPs increased caspase-3, -8, and -9 activities and apoptosis in BECs. ► Phagocytosis played an important role in mediating the effects of LMPs on BECs.

Introduction

Microparticles (MPs) are submicron-sized membranous vesicles (diameters between 0.1 and 1 μm) that contain cell surface proteins and cytoplasmic components and are shed by cells under stress, such as activated and apoptotic cells (Burnier et al., 2009, Hugel et al., 2005). The levels of circulating microparticles, such as leukocyte-derived MPs (LMPs), are significantly increased in patients with pulmonary hypertension (Amabile et al., 2008). Increased levels of pro-coagulant alveolar microparticles are found in alveolar edema samples or bronchoalveolar lavage fluids (BALF) from patients with acute lung injury (ALI), acute respiratory distress syndrome (ARDS), and COPD (Bastarache et al., 2009, Mutschler et al., 2002). Thus, MPs appear to be involved in the pathogenesis of numerous respiratory diseases, particularly those characterized by chronic inflammation.

Damage to bronchial epithelial cells (BECs) that causes ciliary dysfunction is a key property of the pathogenesis of airway inflammation and is thought to be an important contributor to the development of chronic inflammatory pathological changes and airway hyper-responsiveness. A variety of cells types, such as lymphocytes, other leukocytes, platelets, and endothelial cells, can access the bronchi and alveolar spaces. Thus, these cells could be the sources of intra-bronchial and intra-alveolar MPs that accumulate on the surfaces of BECs and function as activating molecules to induce functional changes (Buesing et al., 2011).

There is considerable evidence that substantially increased numbers of circulating LMPs are associated with several pathological conditions of immune and inflammatory disorders, such as rheumatoid arthritis, malaria, septicemia, HIV infection, and preeclampsia (Aupeix et al., 1997, Berckmans et al., 2005, Couper et al., 2010, Meziani et al., 2006, Mostefai et al., 2008). A proteomics analysis of LMPs from malignant lymphocytes identified 413 proteins, including 117 membrane proteins, many of which were associated with various pathologies (Miguet et al., 2006). Thus, LMPs are biomarkers and vectors that carry biological information between cells (Morel et al., 2011).

LMPs derived from human Jurkat T-lymphoma cells are rapidly engulfed by RAW 264.7 mouse macrophages and consequently induce macrophage apoptosis (Distler et al., 2005a). We also observed that LMPs derived from human CEM (human lymphoblastoma) T cells strongly reduced human umbilical vein endothelial proliferation without inducing endothelial cell death (Yang et al., 2008). LMPs also caused a dose-dependent inhibition of Lewis lung carcinoma cells growth, although these effects were associated with increased cellular apoptosis (Yang et al., 2010). Interestingly, resting and cytokine-stimulated human BECs can selectively recognize and engulf apoptotic eosinophils (Sexton et al., 2004, Walsh et al., 1999). However, no studies have investigated the effects of LMPs on BECs with regard to these cells’ phagocytic activities.

Previous studies have also shown that BECS played key roles in immune regulation during the development of bronchial inflammation that was linked to either contact with inflammatory cells or activation by inflammatory mediators in response to pathogen-associated molecular pattern molecules (Bals and Hiemstra, 2004). Monocyte/macrophage-derived microparticles induced increased expression of IL-8 in human bronchial epithelial and alveolar cells (Cerri et al., 2006, Neri et al., 2011). Distler et al. (2005b) confirmed that LMPs induced the expressions of various inflammatory mediators, including IL-6, IL-8, MCP-1, MCP-2, and MMPs, in synovial fibroblasts in rheumatoid arthritis.

Thus, in this study we investigated if human CEM (human lymphoblastoma) T cell-derived microparticles (LMPs) could directly induce apoptosis and cytokine production by human BECs. We also examined if LMPs phagocytosis by BECs played a role in mediating these LMP effects. Our results show that LMPs phagocytosis by BECs is directly related to BECs apoptosis and their production of pro-inflammatory cytokines.

Section snippets

Cell culture

Human bronchial epithelial cells (16HBE) and human lymphoblastoma cells (CEM-T) were purchased from the ATCC (Manassas, VA, USA) and cultured in RPMI-1640 Medium (Gibco BRL, Long Island, NY) supplemented with 10% heat inactivated FBS (Hyclone laboratories, South Logan, UT, USA), 2 mM l-glutamine, 1% nonessential amino acids, 100 units/ml penicillin, and 100 μg/ml streptomycin in a humidified atmosphere of 95% air and 5% CO2 at 37 °C. Cells between passages 4–9 were used.

LMPs generation

LMPs were released into the

Quantifying LMPs released from CEM-T cells

Using differential centrifugation, LMPs were isolated from the supernatants of human CEM T lymphoblastoma cells in the basal state and after stimulation with actinomycin D for 24 h. Identification and quantitation of LMPs used FACS analysis based on size gating and membrane expression of annexin V, a marker of apoptotic cells. Fig. 1A shows the sizes of standard microbeads (blue) on linear scales for SSC and FSC signals, and Fig. 1B shows microbeads fluorescence (blue) on a log-scale vs. SSC on

Discussion

Increasing evidence suggests that microparticles (MPs) that are shed by various cell types under different states of stress can regulate either beneficial or detrimental responses. Lymphocyte-derived microparticles (LMPs) have exhibited both pro- and anti-inflammatory activities in inflammatory and antoimmune diseases. These controversial properties were suggested by previous studies when blood-borne leukocytes were recruited to the airway epithelium for defenses against pathogens in pulmonary

Conclusion

In summary, low concentrations of lymphocytes-derived MPs (LMPs) in the respiratory tract may not affect the viability of BECs under normal physiological conditions. However, under pathological conditions, increasing concentrations of LMPs in bronchi may stimulate the production of pro-inflammatory cytokines by BECs and eventually induce BECs apoptosis through their phagocytosis of LMPs.

Conflict of interest

No conflicts of interest, financial or otherwise, are declared by the authors.

References (42)

  • N. Amabile et al.

    Circulating endothelial microparticle levels predict hemodynamic severity of pulmonary hypertension

    American Journal of Respiratory and Critical Care Medicine

    (2008)
  • K. Aupeix et al.

    The significance of shed membrane particles during programmed cell death in vitro, and in vivo, in HIV-1 infection

    Journal of Clinical Investigation

    (1997)
  • R. Bals et al.

    Innate immunity in the lung: how epithelial cells fight against respiratory pathogens

    European Respiratory Journal

    (2004)
  • J.A. Bastarache et al.

    Procoagulant alveolar microparticles in the lungs of patients with acute respiratory distress syndrome

    American Journal of Physiology: Lung Cellular and Molecular Physiology

    (2009)
  • R.J. Berckmans et al.

    Synovial microparticles from arthritic patients modulate chemokine and cytokine release by synoviocytes

    Arthritis Research and Therapy

    (2005)
  • R.J. Berckmans et al.

    Cell-derived microparticles in synovial fluid from inflamed arthritic joints support coagulation exclusively via a factor VII-dependent mechanism

    Arthritis and Rheumatism

    (2002)
  • E. Boilard et al.

    Platelets amplify inflammation in arthritis via collagen-dependent microparticle production

    Science

    (2010)
  • K.N. Burger

    Greasing membrane fusion and fission machineries

    Traffic

    (2000)
  • L. Burnier et al.

    Cell-derived microparticles in haemostasis and vascular medicine

    Thrombosis and Haemostasis

    (2009)
  • C. Cerri et al.

    Monocyte/macrophage-derived microparticles up-regulate inflammatory mediator synthesis by human airway epithelial cells

    Journal of Immunology

    (2006)
  • K.N. Couper et al.

    Parasite-derived plasma microparticles contribute significantly to malaria infection-induced inflammation through potent macrophage stimulation

    PLOS Pathogens

    (2010)
  • Cited by (16)

    • Activation of NLRP3 inflammasome by lymphocytic microparticles via TLR4 pathway contributes to airway inflammation

      2020, Experimental Cell Research
      Citation Excerpt :

      After 4 h of incubation, cells were fixed with paraformaldehyde, and nuclei were stained with DAPI (Sigma). Fluorescence microscopy was performed as previously described with a Zeiss LSM510 laser scanning confocal microscope attached to a Zeiss Axiovert 200 microscope by using Zeiss Plan-Apo 63 × 1.40 NA oil immersion lens [32]. TLMPs (10 μg/ml) or control was added to cells grown on round cover slips in 24-well plates for 4 h for autophagy marker staining.

    • Dual anti-oxidant and anti-inflammatory actions of the electrophilic cyclooxygenase-2-derived 17-oxo-DHA in lipopolysaccharide- and cigarette smoke-induced inflammation

      2014, Biochimica et Biophysica Acta - General Subjects
      Citation Excerpt :

      TNFα is particularly elevated in the lung of COPD patients [31,48]. Although this cytokine is primarily released by cells of monocytic lineage, production by bronchial epithelial cells has also been reported [31,49–51]. In our experimental model, 16HBE cells did not release detectable amount of TNFα and therefore it was not possible to investigate the action of 17-oxo-DHA on this cell type.

    View all citing articles on Scopus
    View full text