Protective role of mesenchymal stem cells and mesenchymal stem cell-derived exosomes in cigarette smoke-induced mitochondrial dysfunction in mice

https://doi.org/10.1016/j.taap.2019.114788Get rights and content

Highlights

  • The protective role of MSC and EXO in CS-induced lung mitochondrial dysfunction was determined.

  • MSC + EXO treatment showed much protection compared to individual treatments.

  • CS exposure led to significant increase in the mitochondrial fission protein and DAMPs.

  • BEAS2B-MSC co-culture showed protection against the CSE-altered mitochondrial respiration.

  • MSC + EXO treatment protect against CS-mediated against mitochondrial dysfunction.

Abstract

Background

Cigarette smoke (CS)-induced lung inflammation and Chronic Obstructive Pulmonary disease (COPD) involves mitochondrial dysfunction. Mesenchymal stem cells (MSC) and MSC-derived exosomes (EXO) are reported to show therapeutic effects in many animal models of inflammation and injury. In the present study, we determined the role of MSC and EXO intervention in CS-induced lung inflammation with a focus on mitochondrial dysfunction.

Methods

EXO were characterized using Western blot for exosomal markers, tunable resistive pulse sensing by qNano and transmission electron microscopy (TEM). Mitochondrial reporter mice (mt-Keima and mito-QC) were exposed to air or CS for 10 days. mt-Keima mice were treated with intraperitoneal injections of MSC or EXO or MSC and EXO (MSC + EXO) for 10 days. Total cell counts, differential cell counts were performed using automated cell counter and flow cytometry respectively. Further, the levels of pro-inflammatory mediators in bronchoalveolar lavage (BAL) fluid were measured using ELISA. Western blot analysis, quantitative PCR, confocal microscopy were used in the current study to determine the effects in the lungs of CS exposed mice. Seahorse flux analyzer was used to measure the oxidative-phosphorylation (OXPHOS) in the BEAS2B cells and BEAS2B - mMSC co-culture experiments.

Results

CS exposure increased the inflammatory cellular infiltrations in the lungs of the mt-Keima mice. MSC + EXO treatment showed protection compared to individual treatments (MSC or EXO alone). There were no changes in the mitophagy proteins like PINK1 and Parkin, which was also found using the mito-QC mice. CS exposure led to significant increase in the mitochondrial fission protein DRP1 and other DAMPs pathway mediators like S100A4 and S100A8, HMGB1, RAGE and AGE. MSC + EXO treatment increased the gene expression of (fusion genes) mfn1, mfn2 and opa1. Additionally, the rhot1 gene expression was increased in MSC + EXO treatment group compared to Air- and CS exposed groups. BEAS2B-mMSC co-cultures showed protective response against the CSE-altered mitochondrial respiration parameters, confirming the beneficial effect of MSC towards human bronchial lung epithelial cells.

Conclusion

CS affects some of early mitochondrial genes involved in the fission/fusion process, enhancing the damage response along with altered cytokine levels. MSC + EXO combination treatment showed their protective effects. MSC + EXO combination treatment may act against these early events caused by CS exposure owing to its anti-inflammatory and other mitochondrial transfer mechanisms.

Introduction

Cigarette smoke (CS) is the main etiological risk factor involved in the pathogenesis of Chronic Obstructive Pulmonary Disease (COPD) (Churg et al., 2008). CS contains several free radicals and chemicals, which cause lung inflammation (Sundar et al., 2013; Vlahos et al., 2006). Earlier, we have characterized that acute CS exposure causes lung inflammation and changes in some of the early cytokine responses inducing lung damage (Sundar et al., 2017), and CS extract caused mitochondrial dysfunction in lung epithelial cells (Sundar et al., 2019). Mesenchymal stem cells (MSC) and MSC-derived exosomes (EXO) are gaining wide attention as therapeutic intervention in various inflammatory models of disease (D'Agostino et al., 2010; Kennelly et al., 2016; Alipoor et al., 2016; Willis et al., 2018). MSC showed its beneficial effect in several acute lung injury models (Matthay et al., 2010). Further, MSC shows its therapeutic benefits in multitude ways in which transfer of mitochondria is also reported as one of the potential mechanisms (Li et al., 2014). MSCs also exert paracrine effect by releasing the vesicles-like exosomes and other mediators which carry the information and signaling for the cellular protection (Pierro et al., 2013). However, their role in the acute CS-induced lung inflammation and injury is not known. On the other hand exosomal cargo includes but not limited to DNA, mRNA, miRNA, lncRNA and proteins, which play a crucial role in the cell signaling and response (Conigliaro et al., 2017). Exosomes were also reported to carry the functional mitochondrial elements and communicate (Hough et al., 2018).

The role of MSC and exosomes were shown in several cellular and animal models of lung injury (Willis et al., 2018; Matthay et al., 2010; Gupta et al., 2007; Ortiz et al., 2003). Recently, we have shown that plasma-derived extracellular vesicles (EVs) contain distinct protein and miRNA signature among smoker and COPD subjects, compared to never smoking group (Sundar et al., 2019a, Sundar et al., 2019b). Both MSC and EXO were reported to have anti-inflammatory effect in several animal models (Kim et al., 2006). Recently, it was reported that intraperitoneal injection of EXO prevented bronchopulmonary dysplasia (BPD) in rats (Braun et al., 2018). Porzionato et al., found that intratracheal injection of EVs showed better activity compared to MSC in a rat model of BPD (Porzionato et al., 2019). It was also shown that mesenchymal stromal cell exosomes prevent the severity of pulmonary fibrosis induced by bleomycin in mice (Mansouri et al., 2019). Previously, it was reported that CS exposure induces lung inflammation, along with the altered levels of cytokines in the BAL fluid of different mouse strains (Vlahos et al., 2006; Sundar et al., 2017). Keeping in view the beneficial effects of both MSC and EXO, here we investigated the combination effects of bone marrow-derived MSC and EXO (MSC + EXO) in acute CS-induced (10 days) lung inflammation model in mitochondrial reporter mt-Keima mice. We hypothesized that exosomes given along with the MSC have better beneficial outcomes compared to individual treatments (MSC or EXO alone) in acute CS-induced lung inflammation. We have determined the effect of this treatment on 1) inflammatory cytokines/related pathways, 2) key mitochondrial proteins involved in biogenesis, dynamics (fusion and fission process) and mitophagy, and 3) damage-associated molecular patterns (DAMPs).

Section snippets

Materials and methods

All the key biological and/or chemical resources that are used in this study were validated and authenticated (methods and resources) and are of scientific standard from commercial sources.

Isolation and characterization of MSC-derived exosomes

Exosomes were isolated using the Norgen Biotek kit based method and immunoblot analysis was performed to observe for various exosomal markers. The mMSC exosomes were found to be positive for CD63 (using slot-blot analysis), flotillin-1 and Alix as shown in Fig. 1A. The isolated exosomes were not contaminated by the endoplasmic reticulum as the calnexin bands were not detected, but showed positive for the presence of the complex V (mitochondrial OXPHOS component) protein (data not shown). The

Discussion

CS exposure induced mitochondrial gene alterations, structure and function in several types of lung cells in vitro and animal models in vivo as reported previously (Hoffmann et al., 2013; van der Toorn et al., 2007; Ballinger et al., 1996; Hara et al., 2013; Aravamudan et al., 2014). Several of the mitochondrial targeting compounds show beneficial effect in CS-induced lung/cellular dysfunction (Mizumura et al., 2014; Son et al., 2018; Ahmad et al., 2015a). In the present study, we used the

Funding

This study was supported by the NIH R01 HL135613, R21 ES028006, R01 HL133404, and R01 HL137738.

Author contributions

KPM, IKS and IR designed and interpreted the experimental results. KPM and IKS performed all the experiments, analyzed the data and wrote the manuscript. IKS and IR improvised and approved the final version of the manuscript.

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgement

The authors would like to thank Qixin Wang, Ph.D., for his assistance in analyzing the flow data.

References (44)

  • K. Aoshiba et al.

    Oxidative stress, cell death, and other damage to alveolar epithelial cells induced by cigarette smoke

    Tob. Induc. Dis.

    (2003)
  • B. Aravamudan et al.

    Cigarette Smoke-Induced Mitochondrial Fragmentation and Dysfunction in Human Airway Smooth Muscle

    (2014)
  • S.W. Ballinger et al.

    Mitochondrial Genome Damage Associated with Cigarette Smoking

    (1996)
  • N. Bazhanov et al.

    Intraperitoneally infused human mesenchymal stem cells form aggregates with mouse immune cells and attach to peritoneal organs

    Stem Cell Res Ther

    (2016)
  • M.S. Carraway et al.

    Mitochondrial Biogenesis in the Pulmonary Vasculature during Inhalational Lung Injury and Fibrosis

    (2008)
  • A. Churg et al.

    Mechanisms of cigarette smoke-induced COPD: insights from animal models

    Am. J. Physiol. Lung C

    (2008)
  • S.M. Cloonan et al.

    Mitochondria in lung disease

    J. Clin. Invest.

    (2016)
  • A. Conigliaro et al.

    Exosomes: Nanocarriers of biological messages

    Adv. Exp. Med. Biol.

    (2017)
  • B. D’Agostino et al.

    Mesenchymal stem cell therapy for the treatment of chronic obstructive pulmonary disease

    Expert. Opin. Biol. Ther.

    (2010)
  • J.A. Dougherty et al.

    Potential role of Exosomes in mending a broken heart: nanoshuttles propelling future clinical therapeutics forward

    Stem Cells Int.

    (2017)
  • N. Gupta et al.

    Intrapulmonary delivery of bone marrow-derived mesenchymal stem cells improves survival and attenuates endotoxin-induced acute lung injury in mice

    J. Immunol.

    (2007)
  • H. Hara et al.

    Mitochondrial fragmentation in cigarette smoke-induced bronchial epithelial cell senescence

    (2013)
  • Cited by (68)

    View all citing articles on Scopus
    View full text