Alveolar Macrophages As Orchestrators of COPD

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

COPD: Journal of Chronic Obstructive Pulmonary Disease

ISSN: 1541-2555 (Print) 1541-2563 (Online) Journal homepage: https://www.tandfonline.com/loi/icop20

Alveolar Macrophages as Orchestrators of COPD

Prof. Peter J. Barnes

To cite this article: Prof. Peter J. Barnes (2004) Alveolar Macrophages as Orchestrators of
COPD, COPD: Journal of Chronic Obstructive Pulmonary Disease, 1:1, 59-70, DOI: 10.1081/
COPD-120028701

To link to this article: https://doi.org/10.1081/COPD-120028701

Published online: 20 Apr 2004.

Submit your article to this journal

Article views: 2443

View related articles

Citing articles: 36 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=icop20
COPD
Vol. 1, No. 1, pp. 59–70, 2004

BASIC SCIENCE REVIEW

Alveolar Macrophages as Orchestrators of COPD

Peter J. Barnes, D.M., D.Sc., F.R.C.P.

National Heart and Lung Institute, Imperial College, London, UK

ABSTRACT

Alveolar macrophages play a critical role in the pathophysiology of COPD and are a
major target for future anti-inflammatory therapy. Macrophage numbers are markedly
increased in the lung and alveolar space of patients with COPD and are localized to
sites of alveolar destruction. The increased numbers of macrophages may result from
increased recruitment of blood monocytes, prolonged survival in the lung and to a
lesser extent to increased proliferation in the lung. Alveolar macrophages from COPD
patients have an increased baseline and stimulated secretion of inflammatory proteins,
including certain cytokines, chemokines, reactive oxygen species and elastolytic
enzymes, which together could account for all of the pathophysiological features of
COPD. Alveolar macrophages form COPD appear to be resistant to the anti-
inflammatory effects of corticosteriods and this is linked to reduced activity and
expression of histone deacetylase 2, a nuclear enzyme that switches off inflammatory
genes activated through the transcription factor nuclear factor-KB. Alternative anti-
inflammatory therapies that inhibit macrophages are therefore needed in the future to
deal with the chronic inflammation of COPD. These drugs may include resveratrol,
theophylline derivatives, MAP kinase inhibitors and phosphodiesterase-4 inhibitors.

Key Words: Inflammation; Proteases; Macrophage.

INTRODUCTION particles. They also play an important role in the inflam-


matory response and may play a pivotal role in the
Alveolar macrophages play a critical role in innate pathophysiology of COPD. Because of the extraordinary
and acquired immunity, in the defense of the respiratory diversity of macrophage responses they can account for
tract against pathogens and in the clearance of inhaled most of the known features of COPD (1 – 3) (Figure 1).

*Correspondence: Prof. Peter J. Barnes, D.M., D.Sc., F.R.C.P., National Heart and Lung Institute, Imperial College School of
Medicine, Dovehouse St., London SW3 6LY, UK; Fax: 0207-351-5675; E-mail: p.j.barnes@ic.ac.uk.

59

DOI: 10.1081/COPD-120028701 1541-2555 (Print); 1541-2563 (Online)


Copyright D 2004 by Marcel Dekker, Inc. www.dekker.com
ORDER REPRINTS

60 Barnes

Figure 1. Macrophages may play a pivotal role in COPD as they are activated by cigarette smoke extract and secrete many
inflammatory proteins that may orchestrate the inflammatory process in COPD. Neutrophils may be attracted by interleukin(IL)-8,
growth related oncogene-a (GRO-a) and leukotriene B4 (LTB4), monocytes by macrophage chemotactic protein-1 (MCP-1), and
CD8+ lymphocytes by interferon-g inducible protein (IP-10), monokine-induced by interferon-g (Mig) and interferon-inducible
T-cell a-chemoattractant (I-TAC). Release of elastolytic enzymes including matrix metalloproteinases (MMP) and cathepsins
cause elastolysis, and release of transforming growth factor(TGF)-b1 and connective tissue growth factor (CTGF). Release of
TGF-a activates epithelial growth factor receptors (EGFR) which stimulate mucus hypersecretion. Macrophages also generate
reactive oxygen species (ROS) and nitric oxide (NO) which together form peroxynitrite and may contribute to steroid resistance.

INCREASED MACROPHAGE monocytes into the lung, prolonged survival within the
NUMBERS lung or impaired clearance of macrophages from the
respiratory tract.
There is a marked increase (5 –10-fold) in the
numbers of macrophages in airways, lung parenchyma,
BAL fluid and sputum of smokers and patients with Increased Recruitment
COPD. A careful morphometric analysis of macro-
phage numbers in the parenchyma of patients with The increased numbers of macrophages in smokers
emphysema showed a 25-fold increase in the numbers and COPD patients may be explained by increased
of macrophages in the tissue and alveolar space recruitment of monocytes from the circulation in
compared with normal smokers approximately matched response to monocyte-selective chemokines produced
for cigarette exposure (4). Furthermore, macrophages in the lungs. The monocyte-selective chemokine MCP-
are localised to sites of alveolar wall destruction in 1 is increased in sputum and BAL of patients with
patients with emphysema (5,6). Indeed there is a cor- COPD (10,11), with increased expression in alveolar
relation between macrophage numbers in the airways and small airway macrophages (12).
and the severity of COPD (7). CXC chemokines also act as monocyte chemoat-
Clusters of macrophages are also found around small tractants and act via CXCR2. The concentration of the
airways and associated with peribronchiolar fibrosis in CXC chemokine GRO-a which selectively activates
respiratory bronchiolitis (RB) (8). This is exclusively CXCR2 is markedly increased in sputum and BAL of
seen in smokers and ex-smokers and may persist long patients with COPD (10). Monocytes from patients with
after smoking cessation. RB is represented by ill-defined COPD show a greater chemotactic response to GRO-a
parenchymal micronodules detected on high resolution than cells from normal smokers and non-smokers, but
computerized tomography in some cigarette smokers. this is not explained by any increase in CXCR2 density
Long-term follow-up suggests that these nodules may (13). Interestingly, while all monocytes express CCR2,
evolve into centrilobular emphysema and may therefore the receptor for MIP-1, only  30% of monocytes ex-
represent the evolution of emphysema (9). press CXCR2. It is possible that these CXCR2-express-
The increased numbers of macrophages in COPD ing monocytes transform into macrophages that behave
may be explained by increased recruitment of blood differently—e.g. release more inflammatory proteins.
ORDER REPRINTS

Macrophages in COPD 61

Macrophage Proliferation half of the total numbers of macrophages in the lung and
are located within the alveolar walls and in airway walls.
The increased numbers of macrophages in COPD This location may give them an important role in the
may be due to increased recruitment of monocytes regulation of matrix proteins in the lung. Interstitial
from the circulation, but may also be due to increased macrophages may represent an intermediary stage in the
proliferation and prolonged survival in the lungs. maturation of alveolar macrophages from monocytes
Macrophages have a very low proliferation rate in recruited from the pulmonary circulation. Alveolar
the lungs, but we have demonstrated that there is some macrophages are large mature cells with a high cyto-
increase in cell proliferation measured by proliferative plasm/nucleus ratio and resembling other tissue macro-
cell nuclear antigen (PCNA) in macrophages form phages, whereas interstitial macrophages are smaller,
smokers compared to normal and asthmatic macro- more uniform in size, contain few intracytoplasmic
phages (14). Alveolar macrophages also show signs of lamellar inclusions or lysosomes, and more closely
increased proliferation in other chronic inflammatory resemble peripheral blood monocytes. There is increas-
lung diseases (15). ing evidence that alveolar and interstitial macrophages
are distinct cell populations with differing functions and
Macrophage Survival that each population may contribute to inflammatory and
immune responses in the lungs. Alveolar macrophages
Macrophages have a survival time of several months release more inflammatory mediators and show increased
so this is difficult to measure directly. There is a report of chemotaxis, phagocytosis, cytotoxicity, and release of
cigarette particulates in alveolar macrophages over 2 reactive oxygen and nitrogen intermediates than intersti-
years after cessation of smoking (16). In alveolar macro- tial macrophages. Interstitial macrophages express great-
phages from smokers, there is markedly increased er quantities of complement C3 receptor and intercellular
expression of the anti-apoptotic protein Bcl-XL and adhesion molecule-1 (ICAM-1), are more active in
increased expression of p21CIP/WAF1 in the cytoplasm secreting interleukin(IL)-1 and IL-6 and exhibit greater
(14). This may be secondary to oxidative stress which Ia antigen expression, suggesting that they are more
mimics these effects in vitro. This suggests that macro- specialized in immune responses and immunoregulation.
phages may have a prolonged survival in the lungs of They may also have a greater capacity to proliferate
smokers and patients with COPD. On the other hand, compared to alveolar macrophages and this may maintain
acute exposure of rodent and human alveolar macro- the lung macrophage pool. There is a need to further
phages to cigarette smoke extract induces apoptosis, an investigate the differences between alveolar and intersti-
effect that is also mediated via oxidative stress (17). This tial macrophages, particularly in COPD patients; alveolar
suggests that acute and chronic effects of cigarette macrophages may be obtained by bronchoalveolar
smoking on macrophage survival may differ. lavage, whereas interstitial macrophages may be isolated
by digestion of lung parenchyma removed at surgical
Macrophage Clearance resection. There may also be differences between
alveolar macrophages, possibly through different recruit-
Alveolar macrophages are presumed to be cleared ment mechanisms or local factors that may alter their
form the airways predominantly by mucociliary clear- function, but there are currently no surface markers that
ance which is impaired in patients with COPD. Macro- allow separation of subpopulations. Higher density
phages may also be cleared via lymphatic drainage, and alveolar macrophages are smaller and appear less mature
this might also be impaired in COPD patients as a result and may represent the most recently arrived cells. They
of emphysema and fibrosis of small airways. have greater chemotactic and phagocytic activity and
release greater amounts of reactive oxygen species and
inflammatory mediators than lower density cells (20).
MACROPHAGE HETEROGENEITY Alveolar macrophages may also be differentiated by
different antibodies which appear to discriminate a more
There is increasing evidence for distinct populations dendritic phenotype of cell (21).
of macrophages that may play different roles, although
the tools that can differentiate these subpopulations are Dendritic Cells
poorly developed. At least two subpopulations of macro-
phages are found in the lungs (18,19). Alveolar macro- Dendritic cells are a specialized from of macro-
phages are localized to the alveolar spaces and are phage which play a central role in the initiation of the
strategically placed to interact with inhaled pathogens innate and adaptive immune response (22). The
and particles. Interstitial macrophages make up about airways and lungs contain a rich network of dendritic
ORDER REPRINTS

62 Barnes

cells that are localized near the surface, so that they are sputum and exhaled breath (33 – 35), but this is likely to
ideally located to signal the entry of foreign substances be derived predominantly from neutrophils rather than
that are inhaled (23). Dendritic cells can activate a alveolar macrophages. Human alveolar macrophages
variety of other inflammatory and immune cells, in- express cytosolic phospholipase A2 and release LTB4
cluding macrophages, neutrophils, T- and B-lympho- and platelet-activating factor on activation (36). Alveolar
cytes (24). It therefore likely that dendritic cells may macrophages also secrete PGE2 and may contribute to
play an important role in the pulmonary response to the increased PGE2 in exhaled breath of COPD patients
cigarette smoke and other inhaled noxious agents and (35). This is likely to be derived from cyclooxygenase-2
may therefore be a key cellular element in COPD. The (COX-2) which is expressed in macrophages (37). There
mechanisms by which tobacco smoke actives the is increased COX-2 expression in alveolar macrophages
immune system is not yet understood, but a glycopro- from patients with COPD compared to normal control
tein isolated from tobacco has powerful immunostimu- subjects (38).
latory actions (25). There is an increase in the number
of dendritic cells in rat lungs exposed to cigarette
smoke (26) and in the airways and alveolar walls of
smokers (27,28). Pulmonary histiocytosis is a disease Chemokines
caused by dendritic cell granulomata in the lung and is
characterized by destruction of the lung parenchyma Alveolar macrophages have the capacity to release
that resembles emphysema and the adult form of the multiple chemokines leading to the recruitment of sev-
disease occurs almost exclusively in smokers (29,30). eral cell types from the circulation, including monocytes,
In mice exposed to chronic cigarette smoke there is neutrophils and T-lymphocytes.
an increase in dendritic cells in the airways and The best studied chemokine is IL-8 which is
lung parenchyma (31). The role of dendritic cells in released by alveolar macrophages in response to
recruiting other effector cells in COPD deserves fur- several stimuli, including cigarette smoke extract,
ther study. MIP-3a (CCL20) acts on the chemokine endotoxin and IL-1b. Hypoxia also releases IL-8 from
receptor CCR6 that is expressed by immature dendritic alveolar macrophages and this mechanism might be
cells and is potent chemoattractant of dendritic cells. It relevant in severe COPD and exacerbations (39). It is
is expressed by airway epithelial cells in response to likely that macrophages contribute to the high con-
interferon-g (IFN-g) and therefore may be involved in centrations found in the airways of patients with COPD
COPD (32). (40), although neutrophils recruited by macrophage-
derived neutrophil chemokines may be the major
source. Alveolar macrophages from COPD patients
release significantly more IL-8 at baseline and after
SECRETORY ROLE stimulation with IL-1b and cigarette smoke than
macrophages form normal smokers and non-smokers
Alveolar macrophages have the capacity to secrete a and these differences are maintained in cell culture,
very large number of inflammatory mediators, including suggesting that externals factor are not responsible for
lipid mediators, chemokines, cytokines, growth factors this increased activation (41).
and reactive oxygen and nitrogen species. The may Macrophages also have the capacity to release the
therefore play many roles in the respiratory tract and may chemokines IP-10 (CXCL10), I-TAC (CXCL11) and
be pro-inflammatory or anti-inflammatory. They may be Mig (CXCL9), which are chemotactic for CD8+ Tc1
activated by several stimuli, including cigarette smoke, cells via interaction with the CXCR3 receptor ex-
pro-inflammatory cytokines, endotoxin and immune pressed on these cells (42). Macrophages may therefore
stimuli. Overall, alveolar macrophages from COPD play a key role in the recruitment of T-lymphocytes,
patients show greater release of inflammatory mediators and particularly CD8+ cells, to the lungs. The release
than cells from normal smokers, which in turn secrete of interferon-g (IFN-g) by these cells may further re-
more than cells from non-smokers. lease these chemokines, resulting in a chronic inflam-
matory cycle.
Lipid Mediators
Inflammatory Cytokines
Alveolar macrophages have the capacity to generate
leukotrienes (LT) and prostaglandins (PG). In patients Alveolar macrophages release TNF-a in response
with COPD there is an increase in LTB4 concentration in to inflammatory stimuli, including cigarette smoke and
ORDER REPRINTS

Macrophages in COPD 63

may contribute to the elevated concentrations of TNF-a anions (O 2 ) which are converted to hydrogen
in the sputum of COPD patients (40). Granulocyte- peroxide by superoxide dismutase (54). Production of
macrophage colony stimulating factor (GM-CSF) is reactive oxygen species is important for the microbial
also released from macrophages, but unlike chemo- killing function of macrophages, but is also important
kines and TNF-a, its release does not appear to be in activating MAP kinase signal transduction pathways
increased in COPD cells. and the transcription factors NF-kB and AP-1, which in
turn switch on multiple inflammatory genes.
Interleukin-10 Although NO is readily produced by murine alveo-
lar macrophages, it has proved much more difficult to
IL-10 is a potent anti-inflammatory cytokine that is demonstrate NO production and expression of inducible
released from alveolar macrophages in response to NO synthase (iNOS) in human macrophages in vitro.
inflammatory stimuli. IL-10 secretion is markedly However, NO production and iNOS expression is ob-
reduced in alveolar macrophages from patients with served in alveolar macrophages in vivo during pulmonary
asthma (43) and its concentrations are reduced in infections and alveolar NO may play an important role in
sputum of patients with asthma and COPD, suggesting innate immunity of the respiratory tract (55). There is
that a similar abnormality may apply on COPD (44). increased expression of iNOS in alveolar macrophages
IL-10 production appears to be increased in macro- from induced sputum and lung parenchyma in patients
phages from normal smokers (45), but it is not certain with COPD (56 – 58). As NO is produced under con-
whether macrophages from COPD patients show a ditions of oxidative stress in COPD this may lead to
relatively reduced production, as in asthma. increased production of peroxynitrite, accounting for the
increase in 3-nitrotryosine immunoreactivity in alveolar
Growth Factors macrophages from COPD patients (56).

Human alveolar macrophages express transforming


growth factor-b1 (TBF-b1) and TGF-b3 (46). Alveolar REGULATORY MECHANISMS
macrophages form chronic bronchitis patients release
more TGF-b than cells form normal and asthmatic Transcription Factors
subjects (47). Interestingly human alveolar macro-
phages produce less TGF-b than monocytes (48). This The secretion of multiple inflammatory proteins by
might suggest that interstitial and airway macrophages alveolar macrophages is largely a result of increased
are likely to be a more important source of TGF-b than expression of inflammatory genes orchestrated by pro-
alveolar macrophages. Airway macrophages from inflammatory transcription factors, such as NF-kB and
COPD patients show increased expression of TGF-b1 AP-1. There is increased activation of NF-kB in alveolar
and this may contribute to the fibrosis found in small macrophages from COPD patients as evidenced by
airways (49). The increased TGF-b1 by bronchiolar increased nuclear localization of the p65 subunit (59)
epithelial cells of COPD patients may contribute to the and this is further increased during acute exacerbations
chemoattraction of macrophages to the airways and is of the disease (60). NF-kB may play a critical rile in the
correlated with macrophage numbers. increased expression of TNF-a, chemokines such as IL-
Alveolar macrophages produce TGF-a in much 8, GRO-a and MCP-1, GM-CSF, iNOS, proteases such
greater amounts than TGF-b (48) and this may be a as MMP-9 and adhesion molecules such as ICAM-1
major endogenous activator of epidermal growth factor (Figure 2). LPS activates NF-kB in alveolar macro-
(EGF) receptors that play a key role in regulating phages resulting in secretion of GM-CSF and this is
mucus secretion in response to many stimuli, including blocked by an inhibitory of inhibitor of NF-kB kinase-2
cigarette smoke (50). (IKK2) (61).

Reactive Oxygen and Nitrogen Species Signal Transduction Pathways

Macrophages generate reactive oxygen species and The signal transduction pathways involved in
therefore contribute to the increased oxidative stress in macrophages activation and secretion have not been
the airways of patients with COPD (51 – 53). Activating well characterized and it is uncertain how they are
stimuli, such as cigarette smoke, cause activation and affected in COPD. The amplification of inflammatory
assembly of a multicomponent NADPH oxidase in the gene expression in COPD may be linked to abnormal
macrophage cell membrane which generates superoxide activation of signal transduction pathways that regulate
ORDER REPRINTS

64 Barnes

Figure 2. Pivotal role of nuclear factor-kB (NF-kB) in COPD macrophages. Various stimuli activate inhibitor of NF-kB kinase-2
(IKK-2) which causes translocation of NF-kB to the nucleus where it binds to the promoter region of various inflammatory genes,
leading the increased transcription and release of cytokines, chemokines and proteases, and expression of adhesion molecules.

transcription factors, such as NF-kB. Endotoxin psins K, L and S and neutrophil elastase taken up from
activates both the extracellular signal related (ERK) neutrophils (66,67). Alveolar macrophages from
and p38 mitogen-activated protein (MAP) kinase patients with COPD have a greater elastolytic activity
pathways to stimulate expression of cytokine genes at baseline than those from normal smokers and this
(62). PD 098059, an inhibitor of the ERK pathway, is further increased by exposure to cigarette smoke
blocks GM-CSF release induced by LPS, but this is (45,67,68). Macrophages demonstrate this difference
not affected by SB 203580, an inhibitor of the p38 even when maintained in culture for 3 days and there-
pathway. More studies in human alveolar macrophages fore appear to be intrinsically different from the
are required as there appear to be important differences macrophages of normal smokers and non-smoking
in signal transduction pathways between species. There normal control subjects (67). The predominant elasto-
are also important differences in signalling, depending lytic enzyme secreted by alveolar macrophages in
on the activating stimulus and the inflammatory gene COPD patients appears to be MMP-9 (68,69). MMP-9
that is regulated. is also strongly expressed in emphysematous lung at
sites of macrophage accumulation (70). MMP-12
Histone Acetylation (macrophage metalloelastase) appears to play an
important role in cigarette smoke-induced emphysema
Inflammatory genes are regulated by acetylation of in mice and appears to be necessary for release of
core histones around which DNA is wound. Activation activated TNF-a from alveolar macrophages (71). This
of proinflammatory transcription factors, such as NF- then leads to increased expression of E-selectin and
kB and AP-1, results in interaction with co-activator neutrophil accumulation in the lungs. The role of
molecules that have intrinsic histone acetyltransferase MMP-12 in human macrophages is less well defined as
activity (63). The acetylation of histone H4 is involved there is less evidence for its expression in human
in the activation of inflammatory genes (64) and this alveolar macrophages (69,72).
mechanism has been demonstrated in human alveolar Macrophages also release cysteine proteases but
macrophages (65). their role in COPD has not been evaluated. A cysteine
protease inhibitor E-64 significantly reduces the elasto-
lytic activity of alveolar macrophages from COPD pa-
PROTEASES tients, although this component is less marked and less
sustained than MMP-9 secretion (67). Cathepsins K, L
Alveolar macrophages secrete several elastolytic and S have been identified in association with human
enzymes, including MMP-2, MMP-9, MMP-12, cathe- macrophages (66,73 –75).
ORDER REPRINTS

Macrophages in COPD 65

RESPONSES TO DRUGS smokers may be the marked reduction in activity of


histone deacetylase-2 (HDAC2) (65), which is recruited
There have been relatively few studies reporting to activated inflammatory genes by activated gluco-
the effects of drugs on human alveolar macrophages, corticoid receptors to switch of inflammatory genes
particularly in cells derived from COPD patients. Yet (64) (Figure 3). The reduction in HDAC2 activity in
this is important information if these cells play such a macrophages is correlated with increased secretion of
critical role in orchestrating the chronic inflammatory cytokines like TNF-a and IL-8 and reduced response to
process in COPD. corticosteroids. The reduction of HDAC2 activity in
COPD patients may be mediated through oxidative
Corticosteroids stress and peroxynitrite formation (79).

Corticosteroids are ineffective in suppressing Theophylline


inflammation, including cytokines, chemokines and
proteases, in patients with COPD (76 – 78). In vitro Theophylline in low concentrations increases
the release of IL-8, TNF-a and MMP-9 from macro- HDAC activity in alveolar macrophages and in vitro
phages taken from normal subjects and normal smokers reverses the steroid resistance induced by oxidative
is inhibited by corticosteroids, whereas corticosteroids stress (80). This may account for the anti-inflammatory
are ineffective in macrophages from patients with effect of low dose theophylline seen in COPD, in
COPD (41). Curiously, this does not apply to GM-CSF marked contrast to the resistance to corticosteroids
which does not appear to have increased secretion in (81). This suggests that theophylline has the potential
COPD and is suppressed by corticosteroids, albeit to to even unlock the resistance to corticosteroids in the
a lesser extent than in macrophages from normal treatment of COPD (82). In higher concentrations
smokers. The reasons for resistance to corticosteroids theophylline has an inhibitory effect on alveolar
in COPD and to a lesser extent macrophages from macrophages through PDE inhibition (83).

Figure 3. Stimulation of normal alveolar macrophages activates nuclear factor-kB (NF-kB) and other transcription factors to
switch on histone acetyltransferase leading to histone acetylation and subsequently to transcription of genes encoding inflammatory
proteins, such as tumour necrosis factor-a (TNF-a), interleukin-8 (IL-8) and matrix metalloproteinase-9 (MMP-9). Corticosteroids
reverse this by binding to glucocorticoid receptors (GR) and recruiting histone deacetylase-2 (HDAC2). This reverses the histone
acetylation induced by NF-kB and switches off the activated inflammatory genes. In COPD patients cigarette smoke activates
macrophages, as in normal subjects, but oxidative stress (perhaps acting through the formation of peroxynitrite) impairs the activity
of HDAC2. This amplifies the inflammatory response to NF-kB activation, but also reduces the anti-inflammatory effect of
corticosteroids as HDAC2 is now unable to reverse histone acetylation. (View this art in color at www.dekker.com.)
ORDER REPRINTS

66 Barnes

Phosphodiesterase Inhibitors CONCLUSIONS

Several phosphodiesterases (PDE) are expressed in Macrophages are multipotential inflammatory cells
alveolar macrophages, suggesting that PDE inhibitors that play a key role in the defense of the respiratory tract.
may have inhibitory effects on macrophage function. Alveolar macrophages are activated by irritants such as
Alveolar macrophages express PDE 3, 4 and 7A (84). cigarette smoke, leading to a low grade inflammatory
Studies with inhibitors demonstrate that both PDE3 and response through the release of neutrophil, monocyte and
PDE4 inhibitors suppress macrophage activity, but T-lymphocyte chemotactic factors. In COPD this in-
PDE4 inhibitors are less effective than in monocytes flammatory response appears to be greatly amplified,
(83). A clinical trial of a PDE4 inhibitor cilomilast has resulting in excessive inflammation leads to fibrosis of
shown a reduction in the numbers of macro- small airways and release of excessive proteases result-
phages(CD68+ cells) in the airways of COPD patients, ing in destruction of the lung parenchyma. The molecular
presumably through inhibition of monocyte recruitment basis for the amplification of the inflammatory response
into the lungs (85). of macrophages remains to be determined, but may in-
volve impairment of HDAC2 activity as a result of
Resveratrol oxidative and nitrative stress. It is likely that genetic
factors are important in determining this amplification,
Resveratrol, a flavenoid found in red wine, is an although the specific genes involved have not yet been
effective inhibitor of cytokine expression from macro- identified. The alveolar macrophages are resistant to
phages from COPD patients, but its molecular mech- inhibition by corticosteroids, reflecting the poor response
anisms of action have not yet been determined (86). to this treatment in COPD patients. However, other
Resveratrol appears to inhibit NF-kB and AP-1, sug- treatments, such as theophylline, PDE inhibitors and in
gesting that it inhibits the activation of these tran- the future resveratrol and IKK-2 inhibitors may be more
scription factors and thus the expression of multiple effective. Targeting alveolar macrophages is a logical
inflammatory genes, possibly through the activation of approach, since these cells appear to play such a critical
an unknown nuclear receptor. role in orchestrating the chronic inflammatory process in
COPD (92,93).

PHAGOCYTIC FUNCTION
REFERENCES
Macrophages are phagocytic for particles and bacte-
ria and play an important role in host defense. The 1. Shapiro SD. The macrophage in chronic obstruc-
recpeotrs on macrophages that mediate phagocytosis are tive pulmonary disease. Am J Respir Crit Care
largely unknown (87). The macrophage scavenger Med 1999; 160:S29 –S32.
receptor MARCO appears to play an important role in 2. Barnes PJ. Current and future therapies for airway
phagocytosis of bacteria, but its role in COPD gas not yet mucus hypersecretion. Novartis Found Symp 2002;
been explored. The phagocytic potential of macrophages 248:237 –249.
from COPD patients has not been explored, but it is 3. Tetley TD. Macrophages and the pathogenesis of
possible that impaired phagocytosis may result in the COPD. Chest 2002; 121:156S –159S.
increased bacterial load in the respiratory tract of patients 4. Retamales I, Elliott WM, Meshi B, Coxson HO,
with COPD. Pare PD, Sciurba FC, Roger RM, Hayashi S,
Macrophages recognize apoptotic cells via expres- Hogg JC. Amplification of inflammation in em-
sion of phosphatidylserine (PS) which interacts with physema and its association with latent adenoviral
specific receptors on the macrophage surface (88). In- infection. Am J Respir Crit Care Med 2001; 164:
gestion of apoptotic granulocytes by macrophages in- 469 – 473.
duces the secretion of TGF-b1 (89). Neutrophil elastase 5. Finkelstein R, Fraser RS, Ghezzo H, Cosio
cleaves the PS receptor and may thus impair the ability of MG. Alveolar inflammation and its relation to
macrophages to take up apoptotic neutrophils, resulting emphysema in smokers. Am J Respir Crit Care
in increased numbers of apoptotic neutrophils in the Med 1995; 152:1666– 1672.
airways (90). Alveolar macrophages form COPD pa- 6. Meshi B, Vitalis TZ, Ionescu D, Elliott WM, Liu
tients show defective phagocytosis of apoptotic airway C, Wang XD, Hayashi S, Hogg JC. Emphysema-
epithelial cells (91). tous lung destruction by cigarette smoke. The
ORDER REPRINTS

Macrophages in COPD 67

effects of latent adenoviral infection on the lung 17. Aoshiba K, Tamaoki J, Nagai A. Acute cigarette
inflammatory response. Am J Respir Cell Mol Biol smoke exposure induces apoptosis of alveolar
2002; 26:52 – 57. macrophages. Am J Physiol, Lung Cell Mol
7. Di Stefano A, Capelli A, Lusuardi M, Balbo P, Physiol 2001; 281:L1392 –L1401.
Vecchio C, Maestrelli P, Mapp CE, Fabbri LM, 18. Laskin DL, Weinberger B, Laskin JD. Functional
Donner CF, Saetta M. Severity of airflow limita- heterogeneity in liver and lung macrophages. J
tion is associated with severity of airway inflam- Leukoc Biol 2001; 70:163 – 170.
mation in smokers. Am J Respir Crit Care Med 19. Fathi M, Johansson A, Lundborg M, Orre L, Skold
1998; 158:1277– 1285. CM, Camner P. Functional and morphological
8. Fraig M, Shreesha U, Savici D, Katzenstein AL differences between human alveolar and interstitial
Respiratory bronchiolitis: a clinicopathologic study macrophages. Exp Mol Pathol 2001; 70:77 – 82.
in current smokers, ex-smokers, and never-smo- 20. Haugen TS, Nakstad B, Lyberg T. Heterogeneity
kers. Am J Surg Pathol 2002; 26:647 –653. of procoagulant activity and cytokine release in
9. Remy-Jardin M, Edme JL, Boulenguez C, Remy J, subpopulations of alveolar macrophages and
Mastora I, Sobaszek A. Longitudinal follow-up monocytes. INF 1999; 23:15 –23.
study of smoker’s lung with thin-section CT in 21. Spiteri MA, Clarke SW, Poulter LW. Isolation of
correlation with pulmonary function tests. Radiol- phenotypically and functionally distinct macro-
ogy 2002; 222:261 – 270. phage subpopulations from human bronchoalveo-
10. Traves SL, Culpitt S, Russell REK, Barnes PJ, lar lavage. Eur Respir J 1992; 5:717 –726.
Donnelly LE. Elevated levels of the chemokines 22. Banchereau J, Briere F, Caux C, Davoust J,
GRO-a and MCP-1 in sputum samples from Lebecque S, Liu YJ, Pulendran B, Palucka
COPD patients. Thorax 2002; 57:590 – 595. K. Immunobiology of dendritic cells. Annu Rev
11. Capelli A, Di Stefano A, Gnemmi I, Balbo P, Immunol 2000; 18:767 –811.
Cerutti CG, Balbi B, Lusuardi M, Donner 23. Holt PG, Stumbles PA. Regulation of immuno-
CF. Increased MCP-1 and MIP-1b in bronchoal- logic homeostasis in peripheral tissues by dendritic
veolar lavage fluid of chronic bronchitis. Eur cells: the respiratory tract as a paradigm. J Allergy
Respir J 1999; 14:160 – 165. Clin Immunol 2000; 105:421– 429.
12. de Boer WI, Sont JK, van Schadewijk A, Stolk J, 24. Huang Q, Liu D, Majewski P, Schulte LC, Korn
van Krieken JH, Hiemstra PS. Monocyte che- JM, Young RA, et al. The plasticity of dendritic
moattractant protein 1, interleukin 8, and chronic cell responses to pathogens and their components.
airways inflammation in COPD. J Pathol 2000; Science 2001; 294:870– 875.
190:619 –626. 25. Francus T, Klein RF, Staiano-Coico L, Becker CG,
13. Traves SL, Smith SJ, Barnes PJ, Donnelly Siskind GW. Effects of tobacco glycoprotein
LE. Increased migration of monocytes from (TGP) on the immune system. II. TGP stimulates
COPD patients towards GROa is not mediated the proliferation of human T cells and the
by an increase in CXCR2 receptor expression. Am differentiation of human B cells into Ig secreting
J Respir Crit Care Med 2003; 167:A824. cells. J Immunol 1988; 140:1823 – 1829.
14. Tomita K, Caramori G, Lim S, Ito K, Hanazawa T, 26. Zeid NA, Muller HK. Tobacco smoke induced
Oates T, Chiselita I, Jazrawi E, Chung KF, Barnes lung granulomas and tumors: association with
PJ, Adcock IM. Increased p21CIP1/WAF1 and B pulmonary Langerhans cells. Pathology 1995;
cell lymphoma leukemia-xL expression and re- 27:247 – 254.
duced apoptosis in alveolar macrophages from 27. Casolaro MA, Bernaudin JF, Saltini C, Ferrans
smokers. Am J Respir Crit Care Med 2002; VJ, Crystal RG. Accumulation of Langerhans’
166:724 –731. cells on the epithelial surface of the lower res-
15. Bitterman PB, Saltzman LE, Adelberg S, Ferrans piratory tract in normal subjects in association
VJ, Crystal RG. Alveolar macrophage replication. with cigarette smoking. Am Rev Respir Dis 1988;
One mechanism for the expansion of the mononu- 137:406 – 411.
clear phagocyte population in the chronically 28. Soler P, Moreau A, Basset F, Hance AJ. Cigarette
inflamed lung. J Clin Invest 1984; 74:460 – 469. smoking-induced changes in the number and
16. Marques LJ, Teschler H, Guzman J, Costabel differentiated state of pulmonary dendritic cells/
U. Smoker’s lung transplanted to a nonsmoker. Langerhans cells. Am Rev Respir Dis 1989;
Long-term detection of smoker’s macrophages. 139:1112 – 1117.
Am J Respir Crit Care Med 1997; 156:1700– 1702. 29. Tazi A, Soler P, Hance AJ. Adult pulmonary
ORDER REPRINTS

68 Barnes

Langerhans’ cell histiocytosis. Thorax 2000; chronic obstructive pulmonary disease or asthma.
55:405 – 416. Am J Respir Crit Care Med 1996; 153:530– 534.
30. Tazi A, Moreau J, Bergeron A, Dominique S, 41. Culpitt SV, Rogers DF, Shah P, de Matos C,
Hance AJ, Soler P. Evidence that Langerhans cells Russell RE, Donnelly LE, Barnes PJ. Impaired
in adult pulmonary Langerhans cell histiocytosis inhibition by dexamethasone of cytokine release
are mature dendritic cells: importance of the by alveolar macrophages from patients with
cytokine microenvironment. J Immunol 1999; chronic obstructive pulmonary disease. Am J
163:3511– 3515. Respir Crit Care Med 2003; 167:24 – 31.
31. D’Hulst A, Vermeulen KY, Pauwels RA. Cigar- 42. Saetta M, Mariani M, Panina-Bordignon P, Turato
ette smoke exposure causes increase in pulmonary G, Buonsanti C, Baraldo S, Bellettato CM, Papi A,
dendritic cells. Am J Respir Crit Care Med 2002; Corbetta L, Zuin R, Sinigaglia F, Fabbri LM.
164:A604. Increased expression of the chemokine receptor
32. Reibman J, Hsu Y, Chen LC, Bleck B, Gordon CXCR3 and its ligand CXCL10 in peripheral
T. Airway epithelial cells release MIP-3a/CCL20 airways of smokers with chronic obstructive pul-
in response to cytokines and ambient particulate monary disease. Am J Respir Crit Care Med 2002;
matter. Am J Respir Cell Mol Biol 2003; 28:648 – 165:1404 –1409.
654. 43. John M, Lim S, Seybold J, Robichaud A,
33. Woolhouse IS, Bayley DL, Stockley RA. Sputum O’Connor B, Barnes PJ, et al. Inhaled cortico-
chemotactic activity in chronic obstructive pulmo- steroids increase IL-10 but reduce MIP-1a, GM-
nary disease: effect of alpha(1)-antitrypsin defi- CSF and IFN-g release from alveolar macrophages
ciency and the role of leukotriene B(4) and in asthma. Am J Respir Crit Care Med 1998;
interleukin 8. Thorax 2002; 57:709 –714. 157:256– 262.
34. Beeh KM, Kornmann O, Buhl R, Culpitt SV, 44. Takanashi S, Hasegawa Y, Kanehira Y, Yama-
Giembycz MA, Barnes PJ. Neutrophil chemotac- moto K, Fujimoto K, Satoh K, Okamura K. Inter-
tic activity of sputum from patients with COPD: leukin-10 level in sputum is reduced in bronchial
role of interleukin 8 and leukotriene B4. Chest asthma. COPD and in smokers. Eur Respir J 1999;
2003; 123:1240 –1247. 14:309 – 314.
35. Montuschi P, Kharitonov SA, Ciabattoni G, Barnes 45. Lim S, Roche N, Oliver BG, Mattos W, Barnes PJ,
PJ. Exhaled leukotrienes and prostaglandins in Fan CK. Balance of matrix metalloprotease-9 and
COPD. Thorax 2003; 58:585 – 588. tissue inhibitor of metalloprotease-1 from alveolar
36. Shamsuddin M, Chen E, Anderson J, Smith macrophages in cigarette smokers. Regulation by
LJ. Regulation of leukotriene and platelet-activat- interleukin-10. Am J Respir Crit Care Med 2000;
ing factor synthesis in human alveolar macro- 162:1355 –1360.
phages. J Lab Clin Med 1997; 130:615– 626. 46. Coker RK, Laurent GJ, Shahzeidi S, Hernandez-
37. Jiang YJ, Lu B, Choy PC, Hatch GM. Regulation Rodriguez NA, Pantelidis P, du Bois RM, Jeffery
of cytosolic phospholipase A2, cyclooxygenase-1 PK, McAnulty RJ. Diverse cellular TGF-b1 and
and -2 expression by PMA, TNFalpha, LPS and TGF-b3 gene expression in normal human and
M-CSF in human monocytes and macrophages. murine lung. Eur Respir J 1996; 9:2501 – 2507.
Mol Cell Biochem 2003; 246:31 – 38. 47. Vignola AM, Chanez P, Chiappara G, Merendino
38. Taha R, Olivenstein R, Utsumi T, Ernst P, Barnes A, Zinnanti E, Bousquet J, Bellia V, Bonsignore
PJ, Rodger IW, Giaid A. Prostaglandin H synthase G. Release of transforming growth factor-b (TGF-
2 expression in airway cells from patients with b) and fibronectin by alveolar macrophages in
asthma and chronic obstructive pulmonary disease. airway diseases. Clin Exp Immunol 1996;
Am J Respir Crit Care Med 2000; 161:636 – 640. 106:114 –119.
39. Hirani N, Antonicelli F, Strieter RM, Wiesener 48. Toossi Z, Hirsch CS, Hamilton BD, Knuth CK,
MS, Ratcliffe PJ, Haslett C, Donnelly SC. The Friedlander MA, Rich EA. Decreased production
regulation of interleukin-8 by hypoxia in human of TGF-b1 by human alveolar macrophages
macrophages—a potential role in the pathogenesis compared with blood monocytes. J Immunol
of the acute respiratory distress syndrome (ARDS). 1996; 156:3461 – 3468.
Mol Med 2001; 7:685 –697. 49. de Boer WI, van Schadewijk A, Sont JK, Sharma
40. Keatings VM, Collins PD, Scott DM, Barnes HS, Stolk J, Hiemstra PS, van Krieken JH. Trans-
PJ. Differences in interleukin-8 and tumor necro- forming growth factor b1 and recruitment of
sis factor-a in induced sputum from patients with macrophages and mast cells in airways in chronic
ORDER REPRINTS

Macrophages in COPD 69

obstructive pulmonary disease. Am J Respir Crit 60. Caramori G, Romagnoli M, Casolari P, Bellettato
Care Med 1998; 158:1951 –1957. C, Casoni G, Boschetto P, Fan CK, Barnes PJ,
50. Takeyama K, Jung B, Shim JJ, Burgel PR, Dao- Adcock IM, Ciaccia A, Fabbri LM, Pappi
Pick T, Ueki IF, Protin U, Kroschel P, Nadel A. Nuclear localisation of p65 in sputum macro-
JA. Activation of epidermal growth factor recep- phages but not in sputum neutrophils during COPD
tors is responsible for mucin synthesis induced by exacerbations. Thorax 2003; 58:348 – 351.
cigarette smoke. Am J Physiol, Lung Cell Mol 61. Koch A, Giembycz M, Ito K, Lim S, Jazrawi E,
Physiol 2001; 280:L165– L172. Barnes PJ, Adcock I, Erdmann E, Chung
51. Paredi P, Kharitonov SA, Leak D, Ward S, Cramer KF. MAP-kinase modulation of NF-kB-induced
D, Barnes PJ. Exhaled ethane, a marker of lipid GM-CSF release from human alveolar macro-
peroxidation, is elevated in chronic obstructive phages. Am J Respir Cell Mol Biol 2003; in press.
pulmonary disease. Am J Respir Crit Care Med 62. Carter AB, Monick MM, Hunninghake GW. Both
2000; 162:369 – 373. erk and p38 kinases are necessary for cytokine
52. Montuschi P, Collins JV, Ciabattoni G, Lazzeri N, gene transcription. Am J Respir Cell Mol Biol
Corradi M, Kharitonov SA, Barnes PJ. Exhaled 1999; 20:751 – 758.
8-isoprostane as an in vivo biomarker of lung 63. Urnov FD, Wolffe AP. Chromatin remodeling and
oxidative stress in patients with COPD and healthy transcriptional activation: the cast (in order of
smokers. Am J Respir Crit Care Med 2000; appearance). Oncogene 2001; 20:2991 –3006.
162:1175 –1177. 64. Ito K, Barnes PJ, Adcock IM. Glucocorticoid re-
53. Rahman I, van Schadewijk AA, Crowther AJ, ceptor recruitment of histone deacetylase 2 inhibits
Hiemstra PS, Stolk J, Macnee W, de Boer WI. 4- IL-1b-induced histone H4 acetylation on lysines
Hydroxy-2-nonenal, a specific lipid peroxidation 8 and 12. Mol Cell Biol 2000; 20:6891 – 6903.
product, is elevated in lungs of patients with 65. Ito K, Lim S, Caramori G, Chung KF, Barnes PJ,
chronic obstructive pulmonary disease. Am J Adcock IM. Cigarette smoking reduces histone
Respir Crit Care Med 2002; 166:490– 495. deacetylase 2 expression, enhances cytokine ex-
54. Iles KE, Forman HJ. Macrophage signaling and pression and inhibits glucocorticoid actions in alve-
respiratory burst. Immunol Res 2002; 26:95 –105. olar macrophages. FASEB J 2001; 15:1100 –1102.
55. Weinberg JB. Nitric oxide production and nitric 66. Punturieri A, Filippov S, Allen E, Caras I, Murray
oxide synthase type 2 expression by human mono- R, Reddy V, Weiss SJ. Regulation of elastinolytic
nuclear phagocytes: a review. Mol Med 1998; 4: cysteine proteinase activity in normal and cathep-
557 –591. sin K-deficient human macrophages. J Exp Med
56. Ichinose M, Sugiura H, Yamagata S, Koarai A, 2000; 192:789– 800.
Shirato K. Increase in reactive nitrogen species 67. Russell RE, Thorley A, Culpitt SV, Dodd S,
production in chronic obstructive pulmonary Donnelly LE, Demattos C, Fitzgerald M, Barnes
disease airways. Am J Respir Crit Care Med PJ. Alveolar macrophage-mediated elastolysis:
2000; 160:701 – 706. roles of matrix metalloproteinases, cysteine, and
57. van Straaten JF, Postma DS, Coers W, Noordhoek serine proteases. Am J Physiol, Lung Cell Mol
JA, Kauffman HF, Timens W. Macrophages in Physiol 2002; 283:L867 –L873.
lung tissue from patients with pulmonary em- 68. Russell RE, Culpitt SV, De Matos C, Donnelly L,
physema express both inducible and endothelial Smith M, Wiggins J, Barnes PJ. Release and
nitric oxide synthase. Mod Path 1998; 11:648 – activity of matrix metalloproteinase-9 and tissue
655. inhibitor of metalloproteinase-1 by alveolar macro-
58. Maestrelli P, Paska A, Saetta M, Turato G, phages from patients with chronic obstructive
Nowicki Y, Monti S, Formichi B, Miniati M, pulmonary disease. Am J Respir Cell Mol Biol
Fabbri LM. Decreased haem oxygenase-1 and 2002; 26:602 –609.
increased inducible nitric oxide synthase in the 69. Finlay GA, O’Driscoll LR, Russell KJ, D’Arcy
lung of severe COPD patients. Eur Respir J 2003; EM, Masterson JB, FitzGerald MX, O’Connor
21:971 –976. CM. Matrix metalloproteinase expression and
59. DiStefano A, Caramori G, Capelli A, Lusuardi M, production by alveolar macrophages in emphyse-
Gnemmi I, Ioli F, Chung KF, Donner CF, Barnes ma. Am J Respir Crit Care Med 1997; 156:240–
PJ, Adcock IM. Increased expression of NF-kB 247.
in bronchial biopsies from smokers and patients 70. Ohnishi K, Takagi M, Kurokawa Y, Satomi S,
with COPD. Eur Respir J 2002; 20:556 – 563. Konttinen YT. Matrix metalloproteinase-mediated
ORDER REPRINTS

70 Barnes

extracellular matrix protein degradation in human 82. Barnes PJ. Theophylline: new perspectives on
pulmonary emphysema. Lab Invest 1998; an old drug. Am J Respir Crit Care Med 2003;
78:1077– 1087. 167:813 –818.
71. Churg A, Wang RD, Tai H, Wang X, Xie C, Dai J, 83. Dent G, Giembycz MA, Rabe KF, Wolf B, Barnes
Shapiro SD, Wright JL. Macrophage metalloelas- PJ, Magnussen H. Theophylline suppresses human
tase mediates acute cigarette smoke-induced in- alveolar macrophage respiratory burst through
flammation via tumor necrosis factor-a release. phosphodiesterase inhibition. Am J Respir Cell
Am J Respir Crit Care Med 2003; 167:1083 –1089. Mol Biol 1994; 10:565 – 572.
72. Imai K, Dalal SS, Chen ES, Downey R, Schulman 84. Smith SJ, Brookes-Fazakerley S, Donnelly LE,
LL, Ginsburg M, D’Armiento J. Human collage- Barnes PJ, Barnette MS, Giembycz MA. Ubiqui-
nase (matrix metalloproteinase-1) expression in tous expression of phosphodiesterase 7A in
the lungs of patients with emphysema. Am J human proinflammatory and immune cells. Am
Respir Crit Care Med 2001; 163:786 –791. J Physiol, Lung Cell Mol Physiol 2003; 284:
73. Reddy VY, Zhang QY, Weiss SJ. Pericellular L279 – L289.
mobilization of the tissue-destructive cysteine 85. Gamble E, Grootendorst DC, Brightling CE, Troy
proteinases, cathepsins B, L, and S, by human S, Qui Y, Zhu J, Parker D, Vignola AM, Kroegel
monocyte-derived macrophages. Proc Natl Acad C, Morell F, Hansel TT, Rennard SI, Compton C,
Sci U S A 1995; 92:3849 –3853. Ohad A, Tri T, Edelson J, Pavord ID, Rabe KF,
74. Takahashi H, Ishidoh K, Muno D, Ohwada A, Barnes NC, Jeffery PK. Anti-inflammatory effects
Nukiwa T, Kominami E, Kira S. Cathepsin L of the phosphodiesterase 4 inhibitor cilomilast
activity is increased in alveolar macrophages and (Ariflo) in COPD. Am J Respir Crit Care Med
bronchoalveolar lavage fluid of smokers. Am Rev 2003; 168:976-82.
Respir Dis 1993; 147:1562 – 1568. 86. Donnelly LE, Jones GE, Newton R, Barnes
75. Shi GP, Munger JS, Meara JP, Rich DH, Chapman PJ. The anti-inflammatory action of resveratrol
HA. Molecular cloning and expression of human on human airway epithelial cells is not mediated
alveolar macrophage cathepsin S, an elastinolytic via estrogen or glucocorticosteroid receptors. Am J
cysteine protease. J Biol Chem 1992; 267:7258 – Respir Crit Care Med 2003; 165:A614.
7262. 87. Palecanda A, Kobzik L. Receptors for unopso-
76. Keatings VM, Jatakanon A, Worsdell YM, Barnes nized particles: the role of alveolar macrophage
PJ. Effects of inhaled and oral glucocorticoids on scavenger receptors. Curr Mol Med 2001; 1:589 –
inflammatory indices in asthma and COPD. Am J 595.
Respir Crit Care Med 1997; 155:542 –548. 88. Fadok VA, Bratton DL, Rose DM, Pearson A,
77. Culpitt SV, Nightingale JA, Barnes PJ. Effect of Ezekewitz RA, Henson PM. A receptor for
high dose inhaled steroid on cells, cytokines and phosphatidylserine-specific clearance of apoptotic
proteases in induced sputum in chronic obstructive cells. Nature 2000; 405:85 – 90.
pulmonary disease. Am J Respir Crit Care Med 89. Huynh ML, Fadok VA, Henson PM. Phosphati-
1999; 160:1635 –1639. dylserine-dependent ingestion of apoptotic cells
78. Loppow D, Schleiss MB, Kanniess F, Taube C, promotes TGF-beta1 secretion and the resolution
Jorres RA, Magnussen H. In patients with chronic of inflammation. J Clin Invest 2002; 109:41 –50.
bronchitis a four week trial with inhaled steroids 90. Vandivier RW, Fadok VA, Hoffmann PR, Bratton
does not attenuate airway inflammation. Respir DL, Penvari C, Brown KK, Brain JD, Accutso FJ,
Med 2001; 95:115 –121. Henson PM. Elastase-mediated phosphatidylser-
79. Barnes PJ, Ito K, Adcock IM. A mechanism of ine receptor cleavage impairs apoptotic cell
corticosteroid resistance in COPD: inactivation of clearance in cystic fibrosis and bronchiectasis. J
histone deacetylase. Lancet 2003 in press. Clin Invest 2002; 109:661 –670.
80. Ito K, Lim S, Caramori G, Cosio B, Chung KF, 91. Hodge S, Hodge G, Scicchitano R, Reynolds PN,
Adcock IM, Barnes PJ. A molecular mechanism Holmes M. Alveolar macrophages from subjects
of action of theophylline: induction of histone de- with chronic obstructive pulmonary disease are
acetylase activity to decrease inflammatory gene deficient in their ability to phagocytose apoptotic
expression. Proc Natl Acad Sci U.S.A 2002; 99: airway epithelial cells. Immunol Cell Biol 2003;
8921– 8926. 81:289 – 296.
81. Culpitt S, Maziak W, Loukides S, Keller A, Barnes 92. Barnes PJ. New treatments for COPD. Nat Rev,
PJ. Effect of theophylline on induced sputum Drug Discov 2002; 1:437 – 445.
inflammatory indices in COPD patients. Am J 93. Barnes PJ. New treatments for COPD. Thorax
Respir Crit Care Med 1997; 157:A797. 2003; 58:803 –808.
Request Permission or Order Reprints Instantly!

Interested in copying and sharing this article? In most cases, U.S. Copyright
Law requires that you get permission from the article’s rightsholder before
using copyrighted content.

All information and materials found in this article, including but not limited
to text, trademarks, patents, logos, graphics and images (the "Materials"), are
the copyrighted works and other forms of intellectual property of Marcel
Dekker, Inc., or its licensors. All rights not expressly granted are reserved.

Get permission to lawfully reproduce and distribute the Materials or order


reprints quickly and painlessly. Simply click on the "Request Permission/
Order Reprints" link below and follow the instructions. Visit the
U.S. Copyright Office for information on Fair Use limitations of U.S.
copyright law. Please refer to The Association of American Publishers’
(AAP) website for guidelines on Fair Use in the Classroom.

The Materials are for your personal use only and cannot be reformatted,
reposted, resold or distributed by electronic means or otherwise without
permission from Marcel Dekker, Inc. Marcel Dekker, Inc. grants you the
limited right to display the Materials only on your personal computer or
personal wireless device, and to copy and download single copies of such
Materials provided that any copyright, trademark or other notice appearing
on such Materials is also retained by, displayed, copied or downloaded as
part of the Materials and is not removed or obscured, and provided you do
not edit, modify, alter or enhance the Materials. Please refer to our Website
User Agreement for more details.

Request Permission/Order Reprints

Reprints of this article can also be ordered at


http://www.dekker.com/servlet/product/DOI/101081COPD120028701

You might also like