Accepted Manuscript Update on Molecular Mechanisms of Corticosteroid Resistance in Chronic Obstructive Pulmonary Disease Zhilong Jiang, Lei Zhu PII:

S1094-5539(16)30002-5

DOI:

10.1016/j.pupt.2016.01.002

Reference:

YPUPT 1514

To appear in:

Pulmonary Pharmacology & Therapeutics

Received Date: 1 September 2015 Revised Date:

14 January 2016

Accepted Date: 20 January 2016

Please cite this article as: Jiang Z, Zhu L, Update on Molecular Mechanisms of Corticosteroid Resistance in Chronic Obstructive Pulmonary Disease, Pulmonary Pharmacology & Therapeutics (2016), doi: 10.1016/j.pupt.2016.01.002. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

SC

RI PT

ACCEPTED MANUSCRIPT

Obstructive Pulmonary Disease

M AN U

Update on Molecular Mechanisms of Corticosteroid Resistance in Chronic

TE D

Zhilong Jiang*, Lei Zhu* Department of Pulmonary Medicine Zhongshan Hospital, Fudan University No. 180, Feng Lin Road Shanghai 200032 China

AC C

EP

* Corresponding authors: Department of Pulmonary Medicine Zhongshan Hospital, Fudan University No. 180, Feng Lin Road Shanghai, China 200032 Email: [email protected] and [email protected] Tel.: +86 21 64041990 Fax: +86 21 64035399

1

ACCEPTED MANUSCRIPT

Abstract Chronic obstructive pulmonary disease (COPD) is an inflammatory and irreversible pulmonary disorder that is characterized by inflammation and airway destruction. In recent years, COPD has become a global epidemic due to increased air pollution and exposure to cigarette

RI PT

smoke. Current therapeutics using bronchiodialator and anti-inflammatory corticosteroids are most widely used for all patients with persistent COPD, but these approaches are disappointing due to

limited improvement in symptom control and survival rate. More importantly, a certain number of COPD patients are resistant to the corticosteroid treatment and thus their symptoms worsen.

SC

Therefore, more effective anti-inflammatory drugs and combinational treatment are required. Understanding of the underlying molecular and immunological mechanisms is critical to

M AN U

developing new therapeutics. Lung inflammation and the released pro-inflammatory cytokines affect glucocorticoid receptor (GR), histone deacetylase 2 (HDAC2) and surfactant protein D (SP-D) activities in many cell types. Macrophages, neutrophils, airway epithelial cells and lymphocytes are involved in the induction of corticosteroid resistance. This review updated the recent advances in molecular and immunological mechanisms of steroid resistance among patients and animal models with COPD. Meanwhile we discussed novel therapeutic approaches

TE D

in controlling lung inflammation and improving corticosteroid sensitivity among the steroid resistant patients with COPD.

EP

Keywords: Chronic obstructive pulmonary disease (COPD), corticosteroid resistance,

AC C

glucocorticoid receptor (GR), histone deacetylase 2 (HDAC2), surfactant protein D (SP-D)

2

ACCEPTED MANUSCRIPT

Introduction Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory lung disease, characterized by progressive lung tissue destruction, shortness of breath, chronic coughing and mucus production. Recent clinical surveys revealed that COPD accounted for 1.6% of all

RI PT

hospital admission in China and ranked fourth as a leading cause of mortality in urban areas and third in rural areas of China [1]. Due to the increased prevalence and incidence, COPD places a burden on employers and individuals in terms of lost productivity and lost income related to absenteeism, activity limitation, and disability [2]. Cigarette smoke (CS) and biomass fuel use

SC

are major contributors to the high incidence of COPD in China [1]. Other factors include occupational hazards and pathogen infections [3]. Mice with experimental COPD are

M AN U

predisposed to influenza infection. Rhinovirus (RV)-infected mice have high risk of COPD exacerbation [4, 5]. CS-exposed animals develop emphysema, characterized by irreversible alveolar destruction and airspace enlargement [6]. In CS-exposed A549 cells, a large amount of hydrogen peroxide (H2O2) is produced and responsible for cell apoptosis and autophagy [7, 8]. In additional to the debilitating effect on lung epithelial cells, CS acts on alveolar macrophages

TE D

via oxidative stress, under which macrophage phagocytosis activity is largely suppressed [9, 10].

Under oxidative stress, multiple pro-inflammatory cytokines and chemokines such as tumor necrosis factor-alpha (TNF-alpha), interferon-gamma (IFN-gamma), interleukin-1beta (IL-1beta), interleukin-6

(IL-6),

CXCL8,

monocyte

chemoattractant

protein-1

(MCP-1),

matrix

EP

metalloproteinase-12 (MMP-12), macrophage inflammatory protein-2 (MIP-2), MIP-1alpha are increasingly produced from activated macrophages, airway epithelial cells, neutrophils,

AC C

lymphocytes and other cells types. They contribute to the pathogenesis of COPD among patients and animal models [11, 12]. Anti-inflammatory drugs are major therapeutic agents for relieving COPD symptoms. Dexamethasone (Dex) is a synthetic corticosteroid and has been widely used for the treatment of asthma and COPD. However, its effectiveness was recently challenged by developing corticosteroid resistance among 30% of treated patients [13, 14]. A high dose of inhaled or systemic corticosteroid is required for symptom control, but in rare cases, high inhaled doses of steroids fail to control symptoms. Recent research suggests that pro-inflammatory cytokines and other mediators contribute to the development of corticosteroid resistance. In association with increased lung inflammation, the expression and activity of corticosteroid 3

ACCEPTED MANUSCRIPT

receptor (GR), histone deacetylase-2 (HDAC2) and other important molecules are reduced. This review updated the recent advances in molecular and immunological mechanisms of COPD and discussed the novel therapeutic approaches in overcoming corticosteroid resistance during the

RI PT

COPD treatment.

The role of inflammation in COPD

CS exposure creates great damage to lung epithelial cells and activates alveolar macrophages. The CS-exposed lung epithelial cells produce a greater amount of H2O2 and superoxide radicals,

SC

contributing to cell apoptosis [7, 15]. The CS-exposed lung develops emphysema and the lung parenchyma destruction is irreversible even after smoking cessation [7, 14, 16-18]. CS-exposed

M AN U

mice and human subjects have increased bronchoalveolar lavage (BAL) protein levels, hyperplasia of airway epithelial cells and alveolar-capillary barrier permeability [19, 20]. Current smokers have more pro-inflammatory cytokines than ex-smokers [21]. Multiple lung resident cell types such as airway epithelial cells, alveolar macrophages, smooth muscle cells and fibroblasts, are activated and release greater amount of cytokines and chemokines [22-24]. Serum IL-27 and IL-33 levels are elevated in serum, sputum and BAL of COPD patients and

TE D

their expression levels are correlated to disease exacerbation [23, 25, 26]. In addition, IL-33 can up-regulate IL-6 and IL-8 expression in alveolar macrophages in ex vivo [27].

Studies among human COPD samples and mouse models indicate that neutrophils are major

EP

cell infiltrates in the inflamed lung of COPD. Other infiltrates include peripheral monocytes, CD4+ and CD8+ T cell lymphocytes are recruited into the lung and contribute to lung

AC C

inflammation, tissues destruction and remodeling [28]. The activated neutrophils and alveolar macrophages releases multiple pro-inflammatory cytokines, chemokines and other mediators such as myeloperoxidase (MPO), TNF-alpha, IL-1beta, IL-6, IL-17, MCP-1, CCL2, CXCL2, CXCL5, CXCL8 [29, 30]. The increased production of MMP-9 can break down the extracellular matrix of lung tissues into small peptides that are subsequently recognized by T cells for T cell activation and infiltration [31, 32]. Macrophages constitute a heterogeneous cell population, composed of classically activated macrophages (M1 cells) and alternatively activated macrophages (M2 cells). There are altered macrophage phenotypes in the inflamed lung of COPD. Most of them are presented as intermediate phenotypes [33]. A study by Kunz et al 4

ACCEPTED MANUSCRIPT

indicated that the percentage of CD163+ M2 macrophages was increased, but the total number decreased among COPD ex-smokers compared to that of current smokers, indicating that smoking cessation affects macrophage phenotypes and related function [34]. In general, COPD smokers have dominant M2 type macrophages, characterized by high expression of MMP-2,

RI PT

MMP-7 and adenosine A3 receptor (ADORA3) [35]. However information about the dynamic changes and involvement of alveolar macrophage phenotypes is limited in the pathogenesis and inflammation of COPD. More investigation on patients and animal models will address these

SC

issues in the future.

Recent studies also indicated that CD4+ and CD8+ T cells play an important role in the

M AN U

pathogenesis of COPD via releasing IFN-gamma, TNF-alpha, serine protease and granzyme B. COPD exacerbation was inversely associated with the proportion of circulating CD4+ and CD8+ T lymphocytes, but positively associated with the lung T lymphocyte infiltrates. The differences are possibly caused by T cell extravasation from peripheral blood circulation into inflammatory sites and the circulating T cell apoptosis may result in decreases in circulating T cell numbers [36-38]. Hodge et al reported that bronchial brushing-derived CD8+ T cells were increased

TE D

among COPD patients and the expression of TNF-alpha was elevated [39]. In vitro stimulation of the lung derived CD8+ T cells with IL-18 and IL-12 leads to greater production of IFNgamma and TNF-alpha from the stimulated CD8+ T cells [40]. The role of CD8+ T cells in the pathogenesis of COPD is also recently demonstrated in CD8 knock-out mice, in which long-term

EP

exposure to CS did not induce emphysema. Lung inflammation and cytokine expression, such as IFN-gamma-inducible protein-10 (IP-10) and MMP-12 were greatly reduced [41]. A low-dose of

AC C

azithromycin can suppress CD8+ infiltration and airway inflammation in COPD patients [39]. Additional study in ex vivo showed that targeting CD8+ T cells, natural killer (NK) cells and , natural killer T (NKT)-like cells through anti-CD137 antibody can efficiently reduce IFNgamma, TNF-alpha and granzyme B from the isolated peripheral mononuclear cells (PBMC) [42]. Thus T lymphocytes are critically involved in the pathogenesis of COPD. Suppressing T lymphocyte activation and cytokine expression via molecular intervention may have therapeutic potential in COPD control.

5

ACCEPTED MANUSCRIPT

Additional study also indicated that there was increased expression of IL-17A in the infiltrating macrophages, neutrophils, NK cells, NKT cells and gamma delta T-cells after CS exposure [43]. As a major source of IL-17A, Th17 cells also play an important role in lung inflammation. A high number of Th17 cells was observed in the peripheral blood and BAL fluids

RI PT

of COPD patients. The Th17 cell number and IL-17 levels are related to disease severity [44-46]. In contrast, anti-inflammatory T lymphocytes, cytokines and mediators such as regulatory T cells, IL-10 and indoleamine 2,3-dioxygenase (IDO) are reduced among COPD patients [44, 47, 48]. Therefore, COPD progression is associated with increased pro-inflammatory responses, but

SC

reduced anti-inflammatory responses.

M AN U

The role of inflammation in corticosteroid resistance

Inhaled corticosteroids (ICSs) are anti-inflammatory agents and have become major therapeutic agents in symptom control, particularly those with bronchial reversibility. The antiinflammatory effects are thought to be a result of targeting myeloid macrophages among patients with COPD and other chronic inflammatory diseases [49].

TE D

However ICSs have not been shown to prevent disease progression or reduce mortality in clinical trials. Around 30% of the treated patients are low and unresponsive to the steroid treatment. A body of evidence in clinics and animal models has confirmed the involvement of neutrophils, alveolar macrophages, lymphocytes and mast cells in the induction of corticosteroid

EP

resistance [13, 31, 50, 51]. Sputum neutrophils and bronchial CD8+ T cells cannot be reduced in persistent and ex-smokers in patients after short-term steroid treatment [52]. A high dose of ICSs

AC C

has to be administered to partially achieve symptom control [50], but high doses of ICSs result in patient susceptibility to pneumonia, diabetes, dysphonia and other complications [53].

Extensive evidence showed that there are increased amounts of IL-8, MMP-9, phosphoinositide 3-kinase delta, macrophage migration inhibitory factor (MIF) and GR-beta in corticosteroid resistant patients than in steroid sensitive patients. In contrast, the activities of histone deacetylase 2 (HDAC2) [31] and mitogen-activated protein kinase phosphatase 1 [31, 54] are attenuated in steroid resistant patients. In ex vivo experiments showed that Dex failed to suppress lipopolysaccharide (LPS)-induced up-regulation of IL-6, IL-8, GM-CSF, MCP-1 and 6

ACCEPTED MANUSCRIPT

MMP-9 in alveolar macrophages isolated from the steroid resistant patients with COPD [13, 55]. Therefore, not only does inflammation greatly contribute to the pathogenesis of COPD, but it also induces steroid resistance. Neutrophils, lymphocytes and macrophages contribute to the development of steroid resistance. Thus these cells are potential cell targets for molecular

RI PT

intervention in overcoming steroid resistance.

Recent studies indicate that combinational treatment with other agents are more effective in improving steroid sensitivity. CXCR2 antagonists, phosphodiesterase 4 inhibitors (Roflumilast

SC

N-oxide, RNO), p38 mitogen-activating protein kinase inhibitors, and antibodies against IL-1 and IL-17 can increase steroid sensitivity via targeting neutrophils [31, 56]. Other agents such as rosiglitazone,

PPARgamma

agonist

(10-nitro-oleic

acid),

theophylline

and

M AN U

synthetic

phosphoinositide 3-kinase delta inhibitors are also effective in suppressing release of multiple pro-inflammatory cytokines from the cigarette smoke extract (CSE)-exposed macrophages [57, 58]. Resveratrol is an activator of sirtuins, has class III HDAC activity. Pre-incubation of the cultivated airway smooth muscle cells (HASMCs) of COPD with resveratrol can suppress CCL2, IL-6 and IL-8 expression after lipoteichoic acid (LTA)-exposure than pre-incubation with

TE D

dexamethasone. Thus the results provided scientific rationale for the combinational use of antioxidant reagents with corticosteroids for increasing steroid sensitivity in the clinic [22].

The role of glucocorticoid receptor isoforms in corticosteroid resistance

EP

Inflammatory cytokines attenuate steroid sensitivity through modulating multiple protein expression and activity involved in corticosteroid signal pathways. The expression and activity

AC C

of glucocorticoid receptor (GR) are crucial for efficient steroid-mediated anti-inflammatory function. GR is an intracellular receptor for corticosteroid ligand and is coupled with chaperone protein heat shock protein 90 (HSP90) to avoid protein degradation. After GR is bound to the corticosteroid, HSP90 is released to promote GR maturation and binding to glucocorticoid response element (GRE) [59]. Two GR isoforms including GR-alpha and GR-beta exist. GRalpha is abundantly localized in the cytoplasm and suppresses activation of the pro-inflammatory transcription factor NF-kappaB through up-regulation of IKB-alpha and direct binding to NFkappaB. NF-kappaB exerts pro-inflammatory effects through up-regulation of histone acetyltransferase (HAT) and formation of NF-kappaB/HAT complex. After GR-alpha is 7

ACCEPTED MANUSCRIPT

activated via engagement with ligand steroid, GR-alpha can suppress formation of NFkappaB/HAT complex via increasing HDAC2 recruitment to the protein complex, subsequently leading to deacetylation of histones on the promoter region of key pro-inflammatory cytokines (Figure 1) [60, 61]. In contrast, GR-beta is less abundant and can directly bind to GRE with no

RI PT

requirement of ligand [62]. Thus GR-beta is considered an antagonist of GR-alpha and counteracts the anti-inflammatory activity of GR-alpha. In vitro studies have indicated that TNFalpha, IL-1beta, IL-17 and IL-23 can up-regulate GR-beta in PBMCs and other cell types [63, 64]. There is elevated GR-beta expression in the patients infected with respiratory syncytial virus

SC

(RSV) and patients with severe allergic rhinitis [65, 66]. However information about the GR-beta expression in COPD patients is limited, but a few studies showed that expression and activity of

M AN U

GR-alpha was reduced in alveolar epithelial cells and macrophages after exposure to CSE [57, 58]. In addition, GR activity was reduced or lost in CD8+CD28-/- T cells of COPD patients. Meanwhile IFN-gamma and TNF-alpha release were increased from T and NKT-like cells. Thus GR isoform affects steroid sensitivity that can be improved by up-regulation of GR-alpha and down-regulation of GR-beta in alveolar macrophages and T lymphocytes [67].

TE D

The role of GR post- translational modification in corticosteroid resistance The GR activity is critically influenced by post-translational modification. Recent studies have revealed that differential GR post-translational modifications such as phosphorylation, acetylation and small ubiquitin-like modifier 1 (SUMO1) affect GR activities [58, 60, 68, 69].

EP

Thus a balanced GR post-translational modification is critical for effective GR-mediated antiinflammatory effects. Multiple stimuli such as pro-inflammatory cytokines and oxidative stress

AC C

modulate GR activity through post-translational modification.

HSP90 is an important protein for GR stability and maturation. Under physiological conditions, HSP90 and cochaperone p23 are coupled with GR to prevent GR from degradation. However oxidative stress increases HSP90 acetylation and attenuates the protective activity of HSP90. HDAC6 activity is an important deacetylase. Under oxidative stress, HDAC6 activity is reduced and HSP90 acetylation is increased [59, 70, 71]. In addition, GR activity is critically influenced by post-translational phosphorylation. GR phosphorylation at distinct residues affect GR activity differentially [58]. GR phosphorylation at Ser211 residue promotes GR nuclear 8

ACCEPTED MANUSCRIPT

transportation and transcriptional activity; whereas GR phosphorylation at Ser226 and Ser203 residues increase GR nuclear export and suppress nuclear transport [72, 73]. IL-2 and IL-4 are potent inducers of GR phosphorylation. In steroid resistant patients with asthma and Crohn's disease, GR phosphorylation is increased in association with reduced GR binding and nuclear

RI PT

transportation activities in PBMC and epithelial cells. The p38 mitogen-activated kinase (MAPK) and transcription factor 2 are activated [68, 74]. However there is limited information about the effects of GR phosphorylation on steroid resistance in COPD.

SC

In addition to the critical role of GR phosphorylation in GR activity, aberrant GR acetylation affects GR activity and steroid sensitivity. Under physiological conditions, GR

M AN U

activity inversely oscillates in response to diurnal changes of serum glucocorticoid levels. A balanced GR activity is important for antagonizing the biological action of diurnally fluctuating circulating glucocorticoids [75, 76]. Circadian genes Clock and brain and muscle aryl hydrocarbon receptor nuclear translocator-like 1(Bmal1) form protein complex and control the circadian rhythm activity of GR through affecting GR acetylation [77, 78]. In CS-exposed mice, Bmal1 expression and activity were reduced and subsequently decrease GR activity. CXCL5

TE D

expression was elevated from alveolar macrophages and neutrophil infiltrates were increased in the CS-exposed mice. The decreased steroid sensitivity was also observed in Bmal1 knock-out mice after exposure to aerosolized LPS [79]. The silent mating type information regulation 2 homologue 1 (SIRT1), a class III histone deacetylase is potent activator of Bmal1 and other

EP

intracellular mediators. Under oxidative stress, SIRT1 activity is suppressed and correlated with the reduced Bmal1 activity [80]. However, so far there is limited information of altered GR

AC C

acetylation in COPD patients and effect of SIRT1 on GR acetylation and activity.

The role of histone deacetylases (HDACs) in corticosteroid resistance Similar to SIRT1, histone deacetylases (HDACs) are potent deacetylases and critically involved in the biological activities of many transcription factors, receptors and histones. The expression levels and activities of HDACs are critically associated with their pro-and antiinflammatory effects. HDACs are currently divided into class I (1, 2, 3, 8), class II (4, 5, 6, 7, 9, 10) and class IV (11). Different classes of HDACs have distinct activities via epigenetic modification of histone H2A, H2B, H3 and H4 [81]. HDAC7 is pro-inflammatory and promotes 9

ACCEPTED MANUSCRIPT

toll-like receptor 4-dependent IL-12p40 and IL-6 expression [82], whereas HDAC6 and HDAC11 exert pro-inflammatory effects through suppressing anti-inflammatory gene IL-10 expression in antigen presenting cells (APC) [83]. In contrast, HDAC2 and HDAC3 have antiinflammatory properties through suppressing pro-inflammatory gene expression. During pro-

RI PT

inflammatory responses, NF-kappaB is activated and forms protein complex with the CREB binding protein (CBP). Recruitment of HAT promotes histone acetylation and pro-inflammatory gene transcription, such as TNF-alpha, IL-8 and IL-1beta, etc. HDAC2 and HDAC3 counteract with HAT activity through deacetylation of histone on target gene promoter region, and decrease

SC

transcriptional activity via tightening chromatin [13, 60, 81, 84-90]. In addition, HDAC2 promotes phagocytosis activity of alveolar macrophages and facilitates the clearance of invading

M AN U

pathogens [10]. Thus a balanced activity between HAT and HDAC2 is important for effective suppression of inflammation and inflammation resolution [8, 91-94].

A body of evidence has indicated the important role of HDAC2 in the induction of steroid resistance. HDAC2 is greatly reduced in alveolar macrophages of patients with COPD and smokers, the activity is correlated to disease severity and exacerbation [13, 54, 60, 85]. Li M et al

TE D

studies in vitro showed that CSE exposure to human macrophages significantly reduced HDAC2 expression, but increased NF-kappaB-dependent pro-inflammatory cytokine expression such as IL-8 and TNF-alpha [95]. HDAC2 expression was also reduced in PBMCs of patients with COPD and smokers [96]. Oxidative stress is a major factor in HDAC2 reduction. Nitric oxide

EP

and superoxide in macrophages reduce HDAC2 activity through inducing S-nitrotyrosine and aldehyde-adduct modification of HDAC2. Exposure to CSE greatly reduced HDAC2 activity via

AC C

S-nitrotyrosine modification at Cys262 and Cys274 residues in human macrophage-like cell lines [13, 88].

The reduced HADC2 is considered a major factor in the induction of steroid resistance in patients with COPD. A low acetylated GR is required for effective suppression of protein complex formation with NF-kappaB and AP-1. Oxidative stress reduces HDAC2 and GR activities through increasing protein post-translational modification, but it is still elusive whether oxidative stress modulates GR activity through other deacetylase such as SIRT1. Modulation of HDAC2 activity has recently emerged as a major therapeutic strategy in the treatment of 10

ACCEPTED MANUSCRIPT

inflammatory diseases as well as an improvement in steroid sensitivity. Theophylline is a potent HDAC activator and can enhance steroid sensitivity in patients with COPD. Cosio et al reported that a low dose of theophylline treatment significantly suppressed NF-kappaB activity, sputum levels of TNF-alpha, IL-6 and IL-8 in the COPD patients with exacerbation [97]. Alveolar

RI PT

macrophages are major targets for the HDAC2 activator treatment. Sulforaphane is a smallmolecule activator of transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) and can restore steroid sensitivity through increasing HDAC2 activity. Alveolar macrophages isolated from steroid resistant patients with COPD demonstrated greater responses to the steroid

SC

after the treatment with sulforaphane in vitro [13]. The beneficial effects are considered in association with elevated Nrf2 binding to anti-oxidant responsive element (ARE) of anti-oxidant

M AN U

gene and increased expression of anti-oxidant gene proteins such as heme-oxygenase-1 (HO-1) [87, 98]. The anti-oxidant gene protein can increase HDAC2 activity through induction of HDAC2 denitrosylation. A variety of anti-oxidants, such as Chinese herbs, fruits, dietary polyphenols including curcumin, resveratrol, and green tea catechins and quercetin, have been used in the treatment of autoimmune and inflammatory diseases. They are also confirmed effective in increasing steroid sensitivity through restoring HDAC2 activity. For example,

TE D

monocytes treated with curcumin treatment have restored HDAC2 activity after CSE exposure [86]. Thus anti-oxidants have effective therapeutic potential in overcoming steroid resistance among patients with COPD and other inflammatory diseases.

EP

The role of surfactant D in COPD

The role of surfactant protein D (SP-D) in the pathogenesis of COPD and its possible

AC C

involvement in the induction of steroid resistance are not well defined so far. SP-D is a family member of proteins termed collagen-like lectins. It is mainly produced from type II alveolar epithelial cells. Recent studies have revealed that SP-D plays an important role in modulating innate and adaptive immune responses under physiological and pathological conditions. A body of evidence indicated that SP-D can facilitate uptake/binding by alveolar macrophages via interaction with pathogens and cell debris, promote macrophage phagocytosis and facilitate inflammation resolution [99-102]. In addition, SP-D suppresses adaptive immune responses in the inflamed lung tissues to avoid excess inflammation-induced lung tissue injury [103, 104]. Recent observation has revealed that SP-D exerts the immune modulatory effects via interaction 11

ACCEPTED MANUSCRIPT

with dendritic cell (DC)-specific ICAM-grabbing non-integrin (DC-SIGN) and transmembrane receptor signal inhibitory regulatory protein alpha (SIRP-alpha) on antigen presenting cells [99, 105]. The role of SP-D in immune responses is well studied in the asthma animal model, but there is limited information on COPD. SP-D deficient mice developed more severe asthma than

RI PT

wild type mice after the allergen challenge. Th2 cytokines IL-4 and IL-13 were highly produced in the SP-D deficient asthmatic mice [106], indicating the potent role of SP-D in protecting mice from developing Th2 type allergic responses.

SC

Recent studies in clinic and animal models have revealed an elevation of serum SP-D level in COPD and smokers. In contrast, SP-D was reduced in the lung tissues, but it was reversible after

M AN U

inhaled corticosteroid treatment [107-109]. The serum SP-D level is correlated with symptom severity, thus serum SP-D level has been currently considered a risk biomarker for COPD [108]. Multiple factors affect SP-D level, including corticosteroids, TNF-alpha, IL-4, IL-6 and IL-13 [104]. Up-regulation of SP-D by corticosteroid has been demonstrated in vitro and in vivo studies. However SP-D promoter lacks GRE, our previous study has observed that SP-D upregulation by corticosteroids was mediated by GR-tethered c/EBP-beta binding to c/EBP binding

TE D

site at proximal region of SP-D promoter (Jiang Z, et al. Am J Respir Crit Care Med 2014, 198: A2355). In addition, the elevated serum SP-D may be caused by increased alveolar-capillary barrier permeability and leakage of pulmonary SP-D into blood system, due to the increased lung epithelial cells and endothelial cell apoptosis under oxidative stress [19, 110, 111]. It is not

EP

excluded that SP-D may be down regulated in type II alveolar epithelial cells under oxidative stress that was demonstrated in LPS-induced rat lung injury [109], but the reduction of SP-D

AC C

expression cannot be explained merely by altered gene expression and transportation, other mechanisms maybe involved in the process. It is possible that oxidative stress in COPD affects SP-D biological function. A multimeric structure is required for efficient binding of SP-D to cell receptors, however studies in vitro showed that oxidative stress can dissemble multimeric SP-D into a monomeric structure that is not functional and even has pro-inflammatory and chemoattractive property [112]. Thus oxidative stress promotes lung inflammation that may be in association with altered SP-D expression and biological function. Given the role of SP-D in modulating lung inflammation, the reduced pulmonary SP-D may contribute to the induction of steroid resistance not only in COPD, but also in the inflammation-related other lung diseases, 12

ACCEPTED MANUSCRIPT

such as acute lung injury and acute respiratory distress syndrome (ARDS). Modulation of SP-D expression and activity may be a potential therapeutic approach in controlling lung inflammation and improving steroid sensitivity among smokers and patients with COPD.

RI PT

Conclusion and Perspective

In patients with COPD and smokers, oxidative stress activates lung epithelial cells and alveolar macrophages, from which a variety of pro-inflammatory cytokines and other mediators are released. These inflammatory cytokines are potent mediators for the induction of steroid

SC

resistance during steroid therapy by suppressing GR-alpha and HDACs activity. Pulmonary SPD has both an immune protection function against pathogen infection and an immune regulatory

M AN U

property to prevent tissue damage during lung inflammation. However, there are unbalanced levels of SP-D in the inflamed lung and blood stream of patients with COPD and smokers. The altered SP-D expression level potentially causes the development of steroid resistance. Oxidative stress may play an important role in the alteration of SP-D levels via affecting alveolar-capillary barrier permeability, SP-D gene expression and biological function. Anti-oxidants and some transcription factor activators of anti-oxidant genes can suppress variety of pro-inflammatory

TE D

cytokine gene expression, greatly improve steroid sensitivity. The beneficial effects are related to the increased expression and activities of HDACs and GR-alpha in the target immune cells. Thus targeting HDACs and GR-alpha activities and modulation of SP-D expression through molecular intervention have therapeutic potential in overcoming steroid resistance among patients with

EP

COPD.

AC C

Acknowledgements

The work was supported by the National Natural Science Foundation of China to L.Z. (No. 81270137) and Research Grant from Zhongshan Hospital, Fudan University of China to Z.J. We thank Kelly Yiting Jiang for her scientific editing assistance in the preparation of this manuscript.

Disclosure The authors declare no competing financial interests. None of the authors affiliated with this manuscript have any commercial or associations that might pose a conflict interest.

13

ACCEPTED MANUSCRIPT

Declaration of Interest The authors report no conflicts of interest. The author alone are responsible for the content and writing of the paper.

5.

6. 7.

8.

9.

10.

11. 12. 13.

14.

15. 16.

SC

M AN U

4.

TE D

3.

EP

2.

X. Fang, X.Wang, C. et al., COPD in China: the burden and importance of proper management, Chest 139(4) (2011) 920-929. J.G.Patel, S.P. Nagar, et al., Indirect costs in chronic obstructive pulmonary disease: a review of the economic burden on employers and individuals in the United States, International journal of chronic obstructive pulmonary disease 9 (2014) 289-300. D.M. Mannino, A.S. Buist, Global burden of COPD: risk factors, prevalence, and future trends, Lancet 370(9589) (2007) 765-773. A.C. Hsu, A.R. Starkey, et al., Targeting PI3K-p110alpha Suppresses Influenza Virus Infection in Chronic Obstructive Pulmonary Disease, American journal of respiratory and critical care medicine 191(9) (2015) 1012-1023. A. Singanayagam, N. Glanville, et al., A short-term mouse model that reproduces the immunopathological features of rhinovirus-induced exacerbation of COPD, Clinical science 129(3) (2015) 245-258. A.R. Agarwal, F. Yin, Short-term cigarette smoke exposure leads to metabolic alterations in lung alveolar cells, American journal of respiratory cell and molecular biology 51(2) 284-293. T. Goldkorn, S. Filosto, Lung injury and lung cancer caused by cigarette smoke-induced oxidative stress: Molecular mechanisms and therapeutic opportunities involving the ceramide-generating machinery and epidermal growth factor receptor, Antioxid Redox Signal 21(15) (2014) 2149-2174. J.W. Hwang, S. Chung S, et al., Cigarette smoke-induced autophagy is regulated by SIRT1-PARP-1dependent mechanism: implication in pathogenesis of COPD, Arch Biochem Biophys 500(2) (2010) 203-209. Y. Chang, L. Al-Alwan, et al., Genetic deletion of IL-17A reduces cigarette smoke-induced inflammation and alveolar type II cell apoptosis, American journal of physiology Lung cellular and molecular physiology 306(2) (2014) L132-143. N. Noda, K. Matsumoto, et al., Cigarette smoke impairs phagocytosis of apoptotic neutrophils by alveolar macrophages via inhibition of the histone deacetylase/Rac/CD9 pathways, Int Immunol 25(11) (2013) 643-650. G. Caramori, I.M. Adcock, et al., Cytokine inhibition in the treatment of COPD, International journal of chronic obstructive pulmonary disease 9 (2014) 397-412. G. John, K. Kohse, et al., The composition of cigarette smoke determines inflammatory cell recruitment to the lung in COPD mouse models, Clinical science 126(3) (2014) 207-221. D. Malhotra, R.K. Thimmulappa, et al., Denitrosylation of HDAC2 by targeting Nrf2 restores glucocorticosteroid sensitivity in macrophages from COPD patients, J Clin Invest 121(11) (2011) 4289-4302. J.A. Marwick, K. Ito, et al., Oxidative stress and steroid resistance in asthma and COPD: pharmacological manipulation of HDAC-2 as a therapeutic strategy, Expert Opin Ther Targets 11(6) (2007) 745-755. Y. Sun, S. Ito, et al., Acrolein induced both pulmonary inflammation and the death of lung epithelial cells, Toxicol Lett 229(2) (2014) 384-392. I. Rahman, I.M. Adcock, Oxidative stress and redox regulation of lung inflammation in COPD, The European respiratory journal 28(1) (2006) 219-242.

AC C

1.

RI PT

References

14

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

17. M. To, D. Takagi, et al., Sputum plasminogen activator inhibitor-1 elevation by oxidative stressdependent nuclear factor-kappaB activation in COPD, Chest 144(2) (2013) 515-521. 18. M.C. Duan, H.J. Tang, et al., Persistence of Th17/Tc17 cell expression upon smoking cessation in mice with cigarette smoke-induced emphysema, Clinical & developmental immunology 2013 (2013) 350727. 19. A. Nemmar, H. Raza, et al., Evaluation of the pulmonary effects of short-term nose-only cigarette smoke exposure in mice, Exp Biol Med (Maywood) 237(12) (2012) 1449-1456. 20. T. Goldkorn, S. Filosto, Lung injury and cancer: Mechanistic insights into ceramide and EGFR signaling under cigarette smoke, American journal of respiratory cell and molecular biology 43(3) (2010) 259-268. 21. G. Hodge, J. Nairn, et al., Increased intracellular T helper 1 proinflammatory cytokine production in peripheral blood, bronchoalveolar lavage and intraepithelial T cells of COPD subjects, Clinical and experimental immunology 150(1) (2007) 22-29. 22. J. Knobloch, C. Wahl, et al., Resveratrol attenuates the release of inflammatory cytokines from human bronchial smooth muscle cells exposed to lipoteichoic acid in chronic obstructive pulmonary disease, Basic & clinical pharmacology & toxicology 114(2) (2014) 202-209. 23. J. Xia, J. Zhao, et al., Increased IL-33 expression in chronic obstructive pulmonary disease, American journal of physiology Lung cellular and molecular physiology 308(7) (2015) L619-627. 24. D. Li, D. Chen, et al., c-Jun N-terminal kinase and Akt signalling pathways regulating tumour necrosis factor-alpha-induced interleukin-32 expression in human lung fibroblasts: implications in airway inflammation, Immunology 144(2) (2015) 282-290. 25. T. Angata, T. Ishii, et al., Association of serum interleukin-27 with the exacerbation of chronic obstructive pulmonary disease, Physiological reports 2(7) (2014) 26. J. Cao, L. Zhang, et al., IL-27 is elevated in patients with COPD and patients with pulmonary TB and induces human bronchial epithelial cells to produce CXCL10, Chest 141(1) (2012) 121-130. 27. H. Wu, S. Yang, et al., Interleukin-33/ST2 signaling promotes production of interleukin-6 and interleukin-8 in systemic inflammation in cigarette smoke-induced chronic obstructive pulmonary disease mice, Biochemical and biophysical research communications 450(1) (2014) 110-116. 28. M.G. Cosio, J. Majo, et al., Inflammation of the airways and lung parenchyma in COPD: role of T cells, Chest 121(5 Suppl) (2002) 160S-165S. 29. L.L. Shen, Y.N. Liu, et al., Inhalation of glycopyrronium inhibits cigarette smoke-induced acute lung inflammation in a murine model of COPD, International immunopharmacology 18(2) (2014) 358-364. 30. J. Wei, H. Zhao, et al., Bilirubin treatment suppresses pulmonary inflammation in a rat model of smoke-induced emphysema, Biochemical and biophysical research communications 465(2) (2015) 180-187. 31. J. Milara, J. Lluch, et al., Roflumilast N-oxide reverses corticosteroid resistance in neutrophils from patients with chronic obstructive pulmonary disease, The Journal of allergy and clinical immunology 134(2) (2014) 314-322. 32. D.W. Perng, K.C. Su, et al., Long-acting beta2 agonists and corticosteroids restore the reduction of histone deacetylase activity and inhibit H2O2-induced mediator release from alveolar macrophages, Pulm Pharmacol Ther 25(4) (2012) 312-318. 33. P.S. Hiemstra, Altered macrophage function in chronic obstructive pulmonary disease, Annals of the American Thoracic Society 10 Suppl (2013) S180-185. 34. L.I. Kunz, T.S. Lapperre, et al., Smoking status and anti-inflammatory macrophages in bronchoalveolar lavage and induced sputum in COPD, Respiratory research 12 (2011) 34. 35. R. Shaykhiev, A. Krause, et al., Smoking-dependent reprogramming of alveolar macrophage polarization: implication for pathogenesis of chronic obstructive pulmonary disease, Journal of immunology 183(4) (2009) 2867-2883. 36. X. Zhu, A.S. Gadgil, et al., Peripheral T cell functions correlate with the severity of chronic obstructive pulmonary disease, Journal of immunology 182(5) (2009) 3270-3277. 15

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

37. C.M. Freeman, C.H. Martinez, et al., Acute exacerbations of chronic obstructive pulmonary disease are associated with decreased CD4+ & CD8+ T cells and increased growth & differentiation factor15 (GDF-15) in peripheral blood, Respiratory research 16 (2015) 94. 38. S.J. Hodge, G.L. Hodge, et al., Increased production of TGF-beta and apoptosis of T lymphocytes isolated from peripheral blood in COPD, American journal of physiology Lung cellular and molecular physiology 285(2) (2003) L492-499. 39. S. Hodge, G. Hodge G, et al., Increased CD8 T-cell granzyme B in COPD is suppressed by treatment with low-dose azithromycin, Respirology 20(1) (2015) 95-100. 40. C.M. Freeman, M.K. Han, et al., Cytotoxic potential of lung CD8(+) T cells increases with chronic obstructive pulmonary disease severity and with in vitro stimulation by IL-18 or IL-15, Journal of immunology 184(11) (2010) 6504-6513. 41. T. Maeno, A.M. Houghton, et al., CD8+ T Cells are required for inflammation and destruction in cigarette smoke-induced emphysema in mice, Journal of immunology 178(12) (2007) 8090-8096. 42. G. Hodge, M. Holmes, et al., Targeting peripheral blood pro-inflammatory cytotoxic lymphocytes by inhibiting CD137 expression: novel potential treatment for COPD, BMC pulmonary medicine 14 (2014) 85. 43. S. Bozinovski, H.J. Seow, et al., Innate cellular sources of interleukin-17A regulate macrophage accumulation in cigarette- smoke-induced lung inflammation in mice, Clinical science 129(9) (2015) 785-796. 44. K. Maneechotesuwan, K. Kasetsinsombat, et al., Decreased indoleamine 2,3-dioxygenase activity and IL-10/IL-17A ratio in patients with COPD, Thorax 68(4) (2013) 330-337. 45. H. Wang, H. Ying, et al., Imbalance of peripheral blood Th17 and Treg responses in patients with chronic obstructive pulmonary disease, The clinical respiratory journal 2014. 46. H. Solleiro-Villavicencio, R. Quintana-Carrillo, et al., Chronic obstructive pulmonary disease induced by exposure to biomass smoke is associated with a Th2 cytokine production profile, Clinical immunology 161(2) (2015) 150-155. 47. H. Li, Q. Liu, et al., Disruption of th17/treg balance in the sputum of patients with chronic obstructive pulmonary disease, The American journal of the medical sciences 349(5) (2015) 392-397. 48. C.M. Freeman, A.L. McCubbrey, et al., Basal gene expression by lung CD4+ T cells in chronic obstructive pulmonary disease identifies independent molecular correlates of airflow obstruction and emphysema extent, PloS one 9(5) (2014) e96421. 49. U. Baschant, L. Frappart, et al., Glucocorticoid therapy of antigen-induced arthritis depends on the dimerized glucocorticoid receptor in T cells, Proc Natl Acad Sci U S A 108(48) (2011) 19317-19322. 50. C. Dawson, A. Dhanda, et al., NFkappaB and glucocorticoid receptor activity in steroid resistance, J Recept Signal Transduct Res 32(1) (2012) 29-35. 51. E.L. Beckett, R.L. Stevens, et al., A new short-term mouse model of chronic obstructive pulmonary disease identifies a role for mast cell tryptase in pathogenesis, The Journal of allergy and clinical immunology 131(3) (2013) 752-762. 52. S.J. Hoonhorst, N.H. ten Hacken, et al., Steroid resistance in COPD? Overlap and differential antiinflammatory effects in smokers and ex-smokers, PloS one 9(2) (2014) e87443. 53. E. Anton, How and when to use inhaled corticosteroids in chronic obstructive pulmonary disease?, Expert review of respiratory medicine 7(2 Suppl) (2013) 25-32. 54. K. Ito, M. Ito, et al., Decreased histone deacetylase activity in chronic obstructive pulmonary disease, The New England journal of medicine 352(19) (2005) 1967-1976. 55. J. Knobloch, H. Hag, et al., Resveratrol impairs the release of steroid-resistant cytokines from bacterial endotoxin-exposed alveolar macrophages in chronic obstructive pulmonary disease, Basic & clinical pharmacology & toxicology 109(2) (2011) 138-143. 56. P.J. Barnes, Therapeutic approaches to asthma-chronic obstructive pulmonary disease overlap syndromes, The Journal of allergy and clinical immunology 136(3) (2015) 531-545. 57. S.P. Lakshmi, A.T. Reddy, et al., Down-regulated peroxisome proliferator-activated receptor gamma (PPARgamma) in lung epithelial cells promotes a PPARgamma agonist-reversible proinflammatory 16

ACCEPTED MANUSCRIPT

64.

65. 66. 67.

68.

69.

70.

71. 72. 73. 74.

75. 76. 77.

RI PT

63.

SC

62.

M AN U

61.

TE D

60.

EP

59.

AC C

58.

phenotype in chronic obstructive pulmonary disease (COPD), The Journal of biological chemistry 289(10) (2014) 6383-6393. P.J. Barnes, Corticosteroid resistance in patients with asthma and chronic obstructive pulmonary disease. The Journal of allergy and clinical immunology 131(3) (2013) 636-645. J.J. Kovacs, P.J. Murphy, et al., HDAC6 regulates Hsp90 acetylation and chaperone-dependent activation of glucocorticoid receptor, Mol Cell 18(5) (2005) 601-607. K. Ito, S. Yamamura, et al., Histone deacetylase 2-mediated deacetylation of the glucocorticoid receptor enables NF-kappaB suppression, The Journal of experimental medicine 203(1) (2006) 7-13. I.M. Adcock, Glucocorticoid-regulated transcription factors, Pulm Pharmacol Ther 14(3) (2001) 211-219. T. Kino, E. Charmandari, et al., Glucocorticoid receptor: implications for rheumatic diseases, Clin Exp Rheumatol 29(5 Suppl 68) (2011) S32-41. A. Vazquez-Tello, R. Halwani, et al., Glucocorticoid receptor-beta up-regulation and steroid resistance induction by IL-17 and IL-23 cytokine stimulation in peripheral mononuclear cells, J Clin Immunol 33(2) (2013) 466-478. J.C. Webster, R.H. Oakley, et al., Proinflammatory cytokines regulate human glucocorticoid receptor gene expression and lead to the accumulation of the dominant negative beta isoform: a mechanism for the generation of glucocorticoid resistance, Proc Natl Acad Sci U S A 98(12) (2001) 6865-6870. A. Ishida, N. Ohta, et al., Overexpression of glucocorticoid receptor-beta in severe allergic rhinitis, Auris Nasus Larynx 37(5) (2010) 584-588. P.V.Diaz, R.A. Pinto, et al., Increased expression of the glucocorticoid receptor beta in infants with RSV bronchiolitis, Pediatrics 130(4) (2012) e804-811. G. Hodge, H. Jersmann, et al., Lymphocyte senescence in COPD is associated with loss of glucocorticoid receptor expression by pro-inflammatory/cytotoxic lymphocytes, Respiratory research 16 (2015) 2. E. Irusen, J.G. Matthews, et al., p38 Mitogen-activated protein kinase-induced glucocorticoid receptor phosphorylation reduces its activity: role in steroid-insensitive asthma, The Journal of allergy and clinical immunology 109(4) (2002) 649-657. R. Xu, Q. Li, et al., [Acrolein decreases SUMO modification of glucocorticoid receptor and reduces its sensitivity to corticosteroids in airway hypersecretion], Zhonghua yi xue za zhi 92(46) (2012) 3291-3295. M.M. Verheggen, P.T. van Hal, et al., Modulation of glucocorticoid receptor expression in human bronchial epithelial cell lines by IL-1 beta, TNF-alpha and LPS, The European respiratory journal 9(10) (1996) 2036-2043. D.N. Payne, I.M. Adcock, et al., Molecular mechanisms of corticosteroid actions, Paediatr Respir Rev 2(2) (2001) 145-150. W. Chen, T. Dang, et al., Glucocorticoid receptor phosphorylation differentially affects target gene expression, Mol Endocrinol 22(8) (2008) 1754-1766. Z. Wang, J. Frederick, et al., Deciphering the phosphorylation "code" of the glucocorticoid receptor in vivo, The Journal of biological chemistry 277(29) (2002) 26573-26580. H. Bantel, M.L. Schmitz, et al., Critical role of NF-kappaB and stress-activated protein kinases in steroid unresponsiveness, FASEB journal : official publication of the Federation of American Societies for Experimental Biology 16(13) (2002) 1832-1834. E. Charmandari, G.P. Chrousos, et al., Peripheral CLOCK regulates target-tissue glucocorticoid receptor transcriptional activity in a circadian fashion in man, PloS one 6(9) (2011) e25612. T. Kino, Circadian rhythms of glucocorticoid hormone actions in target tissues: potential clinical implications, Sci Signal 5(244) (2012) pt4. D.H. Han, Y.J. Lee, et al., Modulation of glucocorticoid receptor induction properties by core circadian clock proteins, Mol Cell Endocrinol 383(1-2) (2014) 170-180.

17

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

78. N. Nader, G.P. Chrousos, et al., Circadian rhythm transcription factor CLOCK regulates the transcriptional activity of the glucocorticoid receptor by acetylating its hinge region lysine cluster: potential physiological implications, FASEB journal : official publication of the Federation of American Societies for Experimental Biology 23(5) (2009) 1572-1583. 79. J. Gibbs, L. Ince, et al., An epithelial circadian clock controls pulmonary inflammation and glucocorticoid action, Nat Med 20(8) (2014) 919-926. 80. J.W. Hwang, I.K. Sundar, et al., Circadian clock function is disrupted by environmental tobacco/cigarette smoke, leading to lung inflammation and injury via a SIRT1-BMAL1 pathway, FASEB journal : official publication of the Federation of American Societies for Experimental Biology 28(1) (2014) 176-194. 81. A.R. Winkler, K.N. Nocka, et al., Smoke exposure of human macrophages reduces HDAC3 activity, resulting in enhanced inflammatory cytokine production, Pulm Pharmacol Ther 25(4) (2012) 286292. 82. M.R. Shakespear, D.M. Hohenhaus, et al., Histone deacetylase 7 promotes Toll-like receptor 4dependent proinflammatory gene expression in macrophages, The Journal of biological chemistry 288(35) (2013) 25362-25374. 83. F. Cheng, M. Lienlaf, et al., Divergent roles of histone deacetylase 6 (HDAC6) and histone deacetylase 11 (HDAC11) on the transcriptional regulation of IL10 in antigen presenting cells, Mol Immunol 60(1) (2014) 44-53. 84. U. Mahlknecht, J. Will, et al., Histone deacetylase 3, a class I histone deacetylase, suppresses MAPK11-mediated activating transcription factor-2 activation and represses TNF gene expression, Journal of immunology 173(6) (2004) 3979-3990. 85. P.J. Barnes, Role of HDAC2 in the pathophysiology of COPD, Annu Rev Physiol 71 (2009) 451464. 86. K.K. Meja, S. Rajendrasozhan, et al., Curcumin restores corticosteroid function in monocytes exposed to oxidants by maintaining HDAC2. American journal of respiratory cell and molecular biology 39(3) (2008) 312-323. 87. N. Mercado, R. Thimmulappa, et al., Decreased histone deacetylase 2 impairs Nrf2 activation by oxidative stress, Biochemical and biophysical research communications 406(2) (2011) 292-298. 88. S.R. Yang, A.S. Chida, et al., Cigarette smoke induces proinflammatory cytokine release by activation of NF-kappaB and posttranslational modifications of histone deacetylase in macrophages, American journal of physiology Lung cellular and molecular physiology 291(1) (2006) L46-57. 89. D. Adenuga, I. Rahman, Protein kinase CK2-mediated phosphorylation of HDAC2 regulates corepressor formation, deacetylase activity and acetylation of HDAC2 by cigarette smoke and aldehydes, Arch Biochem Biophys 498(1) (2010) 62-73. 90. K. Ito, P.J. Barnes PJ, et al., Glucocorticoid receptor recruitment of histone deacetylase 2 inhibits interleukin-1beta-induced histone H4 acetylation on lysines 8 and 12, Mol Cell Biol 20(18) (2000) 6891-6903. 91. M.D. Cantley, D.P. Fairlie, et al., Inhibiting histone deacetylase 1 suppresses both inflammation and bone loss in arthritis, Rheumatology (Oxford) (2015). 92. B.G. Cosio, B. Mann, et al., Histone acetylase and deacetylase activity in alveolar macrophages and blood mononocytes in asthma, American journal of respiratory and critical care medicine 170(2) (2004) 141-147. 93. S.M. Gao, C.Q. Chen, et al., Histone deacetylases inhibitor sodium butyrate inhibits JAK2/STAT signaling through upregulation of SOCS1 and SOCS3 mediated by HDAC8 inhibition in myeloproliferative neoplasms, Exp Hematol 41(3) (2013) 261-270 e264. 94. S. Vishwakarma, L.R. Iyer, et al., Tubastatin, a selective histone deacetylase 6 inhibitor shows antiinflammatory and anti-rheumatic effects, International immunopharmacology 16(1) (2013) 72-78. 95. M. Li, X. Zhong, et al., Effect of erythromycin on cigarette-induced histone deacetylase protein expression and nuclear factor-kappaB activity in human macrophages in vitro, International immunopharmacology 12(4) (2012) 643-650. 18

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

96. Y. Chen, P. Huang, et al., Histone deacetylase activity is decreased in peripheral blood monocytes in patients with COPD, J Inflamm (Lond) 9 (2012) 10. 97. B.G. Cosio, A. Iglesias,et al., Low-dose theophylline enhances the anti-inflammatory effects of steroids during exacerbations of COPD, Thorax 64(5) (2009) 424-429. 98. Y. Son, J.H. Lee, et al., Therapeutic roles of heme oxygenase-1 in metabolic diseases: curcumin and resveratrol analogues as possible inducers of heme oxygenase-1, Oxid Med Cell Longev 2013 (2013) 639541. 99. W.J. Janssen, K.A. McPhillips, et al., Surfactant proteins A and D suppress alveolar macrophage phagocytosis via interaction with SIRP alpha, American journal of respiratory and critical care medicine 178(2) (2008) 158-167. 100. K.A. Soltysiak, E.J. van Schaik, Surfactant Protein D Binds to Coxiella burnetii and Results in a Decrease in Interactions with Murine Alveolar Macrophages, PloS one 10(9) (2015) e0136699. 101. S. Geunes-Boyer, T.N. Oliver, et al., Surfactant protein D increases phagocytosis of hypocapsular Cryptococcus neoformans by murine macrophages and enhances fungal survival, Infection and immunity 77(7) (2009) 2783-2794. 102. C. Winkler, K. Huper, et al., Surfactant protein D modulates pulmonary clearance of pollen starch granules, Experimental lung research 36(9) (2010) 522-530. 103. A. Haczku, Y. Cao, et al., IL-4 and IL-13 form a negative feedback circuit with surfactant protein-D in the allergic airway response, Journal of immunology 176(6) (2006) 3557-3565. 104. L. Hortobagyi, S. Kierstein, et al., Surfactant protein D inhibits TNF-alpha production by macrophages and dendritic cells in mice, The Journal of allergy and clinical immunology 122(3) (2008) 521-528. 105. C.F. Liu, M. Rivere, et al., Surfactant protein D inhibits mite-induced alveolar macrophage and dendritic cell activations through TLR signalling and DC-SIGN expression, Clin Exp Allergy 40(1) (2010) 111-122. 106. B. Schaub, R.M. Westlake, et al., Surfactant protein D deficiency influences allergic immune responses, Clin Exp Allergy 34(12) (2004) 1819-1826. 107. M.W. Sims, R.M. Tal-Singer, et al., Chronic obstructive pulmonary disease and inhaled steroids alter surfactant protein D (SP-D) levels: a cross-sectional study, Respiratory research 9 (2008) 13. 108. B.A. Ozyurek, S.S. Ulasli, et al., Value of serum and induced sputum surfactant protein-D in chronic obstructive pulmonary disease, Multidiscip Respir Med 8(1) (2013) 36. 109. L.H. Shu, X.D. Xue, et al., Effect of dexamethasone on the content of pulmonary surfactant protein D in young rats with acute lung injury induced by lipopolysaccharide, Zhongguo Dang Dai Er Ke Za Zhi 9(2) (2007) 155-158. 110. D. Behera, T. Balamugesh, et al., Serum surfactant protein-A levels in chronic bronchitis and its relation to smoking, Indian J Chest Dis Allied Sci 47(1) (2005) 13-17. 111. T. Iizuka, Y. Ishii, et al., Nrf2-deficient mice are highly susceptible to cigarette smoke-induced emphysema, Genes Cells 10(12) (2005) 1113-1125. 112. E.N. Atochina-Vasserman, S-nitrosylation of surfactant protein D as a modulator of pulmonary inflammation, Biochim Biophys Acta 1820(6) (2012) 763-769.

Legends

Figure 1. Schematic diagram of steroid resistance in the patients with COPD and smokers. Cigarette smoke exposure and pro-inflammatory stimuli activate p38MAPK, but attenuate HDAC2 expression and activity in alveolar macrophages and epithelial cells through oxidative stress. The GR-alpha expression and activity are reduced, but GR-beta expression is enhanced, 19

ACCEPTED MANUSCRIPT

lead to enhanced activation of NF-kappaBp65 and unresponsiveness to corticosteroid. The attenuated GR-alpha activity negatively affects transactivation and transrepression of steroid through different signaling pathways: complex protein tethered GR-alpha indirect binding and

Abbreviations Acute respiratory distress syndrome (ARDS) Adenosine A3 receptor (ADORA3) Airway smooth muscle cells (HASMCs)

Antigen presenting cells (APC) Anti-oxidant responsive element (ARE)

M AN U

Alternatively activated macrophages (M2 cells)

SC

RI PT

direct GR-alpha binding to GRE of target gene promoters.

Brain and muscle aryl hydrocarbon receptor nuclear translocator-like 1(Bmal1) Bronchoalveolar lavage (BAL)

Chronic obstructive pulmonary disease (COPD)

TE D

Cigarette smoke (CS) Cigarette smoke extract (CSE)

Classically activated macrophages (M1 cells) Corticosteroid receptor (GR)

EP

CREB binding protein (CBP)

Dendritic cell (DC)-specific ICAM-grabbing non-integrin (DC-SIGN)

AC C

Dexamethasone (Dex)

Glucocorticoid response element (GRE) Heme-oxygenase-1 (HO-1)

Histone acetyltransferase (HAT) Histone deacetylase-2 (HDAC2) Hydrogen peroxide (H2O2) IFN-gamma-inducible protein-10 (IP-10) Indoleamine 2,3-dioxygenase (IDO) 20

ACCEPTED MANUSCRIPT

Inhaled corticosteroids (ICSs) Interferon-gamma (IFN-gamma) Interleukin-1beta (IL-1beta)

RI PT

Interleukin-6 (IL-6) Lipopolysaccharide (LPS) Lipoteichoic acid (LTA) Macrophage inflammatory protein-2 (MIP-2)

SC

Matrix metalloproteinase-12 (MMP-12) Monocyte chemoattractant protein-1 (MCP-1)

Natural killer (NK) cells Natural killer T (NKT) cells

M AN U

Myeloperoxidase (MPO)

Nuclear factor erythroid 2-related factor 2 (Nrf2) p38 mitogen-activated kinase (p38MAPK) Peripheral mononuclear cells (PBMC)

Rhinovirus (RV)

TE D

Respiratory syncytial virus (RSV)

Roflumilast N-oxide (RNO)

EP

Shock protein 90 (HSP90)

Signal inhibitory regulatory protein alpha (SIRP-alpha)

AC C

Silent mating type information regulation 2 homologue 1 (SIRT1) Small ubiquitin-like modifier 1 (SUMO1) Surfactant protein D (SP-D)

Tumor necrosis factor-alpha (TNF-alpha)

21

ACCEPTED MANUSCRIPT

RI PT

Cigarette smoking (CS) Pro-inflammatory stimuli Corticosteroid (GC)

p38 MAPK

SC

HDAC2 Acetyl

M AN U

GRα

p65

IkBα

IL-10 IL-1Ra

TE D

+

HDAC2

-

Co-activator HAT

+

TSLP

AC C

SP-D

c/EBP

CXCL5 GM-CSF

p65

EP

Co-activator HAT

GRβ

p65

STATs

Co-activator HAT

Co-activator HAT

Figure 1

Update on molecular mechanisms of corticosteroid resistance in chronic obstructive pulmonary disease.

Chronic obstructive pulmonary disease (COPD) is an inflammatory and irreversible pulmonary disorder that is characterized by inflammation and airway d...
565B Sizes 0 Downloads 5 Views