You are on page 1of 9

Therapeutic Advances in Respiratory Disease

http://tar.sagepub.com/

COPD: a multifactorial systemic disease


Alice Huertas and Paolo Palange
Ther Adv Respir Dis 2011 5: 217 originally published online 23 March 2011
DOI: 10.1177/1753465811400490

The online version of this article can be found at:


http://tar.sagepub.com/content/5/3/217

Published by:

http://www.sagepublications.com

Additional services and information for Therapeutic Advances in Respiratory Disease can be found at:

Email Alerts: http://tar.sagepub.com/cgi/alerts

Subscriptions: http://tar.sagepub.com/subscriptions

Reprints: http://www.sagepub.com/journalsReprints.nav

Permissions: http://www.sagepub.com/journalsPermissions.nav

Citations: http://tar.sagepub.com/content/5/3/217.refs.html

>> Version of Record - May 24, 2011

OnlineFirst Version of Record - Mar 23, 2011

What is This?

Downloaded from tar.sagepub.com at UNIV OF LETHBRIDGE on August 19, 2014


Therapeutic Advances in Respiratory Disease Review

Ther Adv Respir Dis


COPD: a multifactorial systemic disease (2011) 5(3) 217—224
DOI: 10.1177/
1753465811400490
Alice Huertas and Paolo Palange
! The Author(s), 2011.
Reprints and permissions:
http://www.sagepub.co.uk/
Abstract: Chronic obstructive pulmonary disease (COPD) has traditionally been considered a journalsPermissions.nav
disease of the lungs secondary to cigarette smoking and characterized by airflow obstruction
due to abnormalities of both airway (bronchitis) and lung parenchyma (emphysema). It is now
well known that COPD is associated with significant systemic abnormalities, such as renal and
hormonal abnormalities, malnutrition, muscle wasting, osteoporosis, and anemia. However, it
is still unclear whether they represent consequences of the pulmonary disorder, or whether
COPD should be considered as a systemic disease. These systemic abnormalities have been
attributed to an increased level of systemic inflammation. Chronic inflammation, however, may
not be the only cause of the systemic effects of COPD. Recent data from humans and animal
models support the view that emphysema may be a vascular disease. Other studies have
highlighted the role of repair failure, bone marrow abnormality, genetic and epigenetic factors,
immunological disorders and infections as potential causes of COPD systemic manifestations.
Based on this new evidence, it is reasonable to consider COPD, and emphysema in particular,
as ‘a disease with a significant systemic component’ if not a ‘systemic disease’ per se. The aim
of this review is to give an overview of the most relevant and innovative hypothesis about the
extrapulmonary manifestations of COPD.

Keywords: chronic obstructive pulmonary disease, inflammation, oxidative stress, systemic


manifestations

Introduction the last two decades, including renal and hor- Correspondence to:
Paolo Palange, MD
The American Thoracic Society and the monal abnormalities [Palange, 1998], muscle Department of Public
European Respiratory Society define chronic wasting [Remels et al. 2007], osteoporosis Health and Infectious
Diseases, Sapienza
obstructive pulmonary disease (COPD) as ‘a pre- [Bolton et al. 2004], anemia [John et al. 2005] University of Rome, Rome,
ventable and treatable disease state characterized and reduction in circulating bone marrow pro- Italy
paolo.palange@
by airflow limitation that is not fully reversible. genitors [Palange et al. 2006]. Although these uniroma1.it
The airflow limitation is usually progressive and systemic manifestations have been described for Alice Huertas, MD
is associated with an abnormal inflammatory years in COPD patients, it is still unclear whether Department of Public
Health and Infectious
response of the lungs to noxious particles or they represent consequences of the pulmonary Diseases, Sapienza
gases, primarily caused by cigarette smoking. disorder, or whether COPD should be consid- University of Rome, Rome,
Italy
Although COPD affects the lungs, it also pro- ered as a systemic disease. The importance of INSERM U999 ‘‘Pulmonary
duces significant systemic consequences’ [Celli establishing the distinction between a respiratory Hypertension:
Pathophysiology and Novel
and MacNee, ATS/ERS Task Force, 2004]. It is disease with extrapulmonary manifestations and Therapies’’ Centre
interesting to point out how the definition of a systemic inflammatory state with multiple com- Chirurgical Marie
Lannelongue, Le Plessis-
COPD has evolved including the ‘systemic con- promised organs is justified by different thera- Robinson, France;
sequences’ of the disease. peutic options: in the first definition, therapy is University Paris-Sud 11,
Orsay, France
primarily centered on the lungs, whereas in the
The natural history of the disease reveals numer- second, therapy could aim at the systemic inflam-
ous extrapulmonary manifestations and comor- matory state.
bidity factors that complicate the evolution of
COPD, thereby altering the prognosis and qual- Furthermore, since COPD represents a group of
ity of life of patients [Barnes and Celli, 2009; pulmonary abnormalities of either airways (bron-
Agusti and Soriano, 2008]. Many extrapulmon- chitis) or lung parenchyma (emphysema), it is
ary effects of COPD have been described over reasonable to raise the question: does the

http://tar.sagepub.com 217

Downloaded from tar.sagepub.com at UNIV OF LETHBRIDGE on August 19, 2014


Therapeutic Advances in Respiratory Disease 5 (3)

systemic involvement of the disease vary depend- loss and low body mass index (BMI) are associ-
ing on the etiology and pathophysiology of each ated with increased mortality [Schols et al. 1998;
of the two different COPD phenotypes? Gray-Donald et al. 1996]. Interestingly, a ran-
domized, controlled trial showed that nutritional
The aim of this review is to give an overview of supplementation resulted in improved exercise
the most relevant and innovative hypotheses capacity in well nourished, but not in undernour-
regarding the extrapulmonary manifestations of ished patients [Steiner et al. 2003]. It is also
COPD. known that patients with low BMI have higher
levels of inflammation than normal BMI patients
Systemic inflammation [Huertas et al. 2010]. On the other hand, it has
Among the numerous extrapulmonary effects of been shown that patients who have relative anor-
COPD, systemic inflammation has been widely exia and high levels of inflammation are least
studied and considered as an important key likely to respond to nutritional supplementation
between the pulmonary disease and the related [Creutzberg et al. 2000]. Therefore, it is reason-
systemic manifestations. Many studies have able to wonder whether the inflammation causes
reported changes in various inflammatory cells the reduction in BMI, whether the low BMI
and mediators, including neutrophils, lympho- induces a higher level of inflammation, or
cytes, acute-phase reactants, and cytokines. whether a factor causing both inflammation and
Gan and colleagues recently performed a meta- low BMI could exist in these patients.
analysis that showed systemic inflammation is pre- Understanding this aspect is important in order
sent during COPD exacerbations and stable to choose whether to treat the inflammation or
phases of the disease: increased numbers of leuko- the outcome.
cytes, levels of acute-phase response proteins
(C-reactive protein and fibrinogen), cytokines Vascular abnormalities
such as interleukin (IL)-6, and tumor necrosis The loss of alveolar capillary endothelial cells has
factor (TNF)-a are present in the peripheral been observed in emphysema for almost 50 years
blood of COPD patients [Gan et al. 2004]. [Liebow, 1959], but new data derived from inves-
Systemic inflammation has been implicated in tigations on the vascular nature of emphysema
the pathogenesis of the majority of COPD sys- suggest a systemic involvement of the disease
temic effects, including weight loss [Wouters, [Voelkel and Taraseviciene-Stewart, 2005; Santos
2002], skeletal muscle dysfunction [Langen et al. et al. 2002; Shapiro, 2000; Yamato et al. 1996].
2001], cardiovascular diseases [Sin and Man, The vascular endothelial growth factor (VEGF)
2003], and osteoporosis [Biskobing, 2002], plays an important role in the vascular pathogen-
although it is still controversial whether this so- esis of COPD, particularly in emphysema. The
called low-grade systemic inflammation represents VEGF has a fundamental role in physiological
the consequence of pulmonary inflammation into and pathophysiological angiogenesis and regula-
the systemic vascular bed [Agustı̀ et al. 2003], or tion of endothelial cell differentiation. This has
whether it is a systemic inflammation. Vernooy been demonstrated by the lethality of VEGF
and colleagues, as well as Hurst and colleagues, knockout mice and by the abnormal vasculogen-
failed to show a relationship between TNF-a and esis of the heart and large vessels following the loss
IL-8 values in induced sputum and plasma, sug- of a single copy of the VEGF gene [Carmeliet et al.
gesting that the systemic inflammation in COPD 1996]. The VEGF, also known as the vascular per-
is not linked to the pulmonary inflammation in meability factor, displays several key functions in
these patients [Hurst et al. 2005; Vernooy et al. the lung, such as migration and proliferation of
2002]. Although inflammation is certainly one of endothelial cells, monocyte adhesion, angiogene-
the major features of COPD, we still need to sis, vasodilatation, and enhanced permeability, as
understand whether the local inflammation is suf- well as early hemangioblast development. Above
ficient to induce systemic effects, or whether a all, a crucial role in the genesis of emphysema
second pathogenetic event is required. has been developed based on the observation
Therefore, further studies are needed to elucidate that blockade of VEGF receptor (VEGFR) -1
the origin of the systemic inflammation in COPD. and -2 arrests lung growth and leads to an emphy-
sematous phenotype in a murine model
Malnutrition [McGrath-Morrow et al. 2005]. Therefore, an
Several studies have demonstrated an association interesting model of VEGFR blockade with
between poor nutrition and COPD, where weight SU5416 results in endothelial cell apoptosis

218 http://tar.sagepub.com

Downloaded from tar.sagepub.com at UNIV OF LETHBRIDGE on August 19, 2014


A Huertas and P Palange

causing apoptosis-dependent emphysema in prevention of repair of lung injury. In addition,


rodents [Tang et al. 2004; Kasahara et al. 2000]. cigarette smoke can kill endothelial cells and
endothelial cell precursors [Hoshino et al.
Alveolar maintenance is defined as the preserva- 2005], and inhibit airway epithelial cell chemo-
tion of the alveolar gas-exchange area by limiting taxis and proliferation [Wang et al. 2001].
the destruction of alveoli and airspace enlarge- Abnormal mesenchymal repair functions have
ment, the hallmark of emphysema. The finding been observed in fibroblasts obtained from
that decreased VEGF or VEGF signaling causes emphysematous human lung. Holz and col-
experimental emphysema leads to the concept leagues have demonstrated that fibroblasts cul-
that alveolar maintenance is required for struc- tured from lungs of emphysematous patients
tural preservation of the lung. This structure, proliferate more slowly than fibroblasts obtained
when disrupted by cigarette smoke, for example, from age-matched control lungs [Holz et al.
causes emphysema [Taraseviciene-Stewart and 2004]. Rennard and colleagues have extended
Voelkel, 2008; Tuder and Voelkel, 2001]. these results to demonstrate that fibroblasts
Therefore, it appears quite legitimate to raise from emphysematous patients are also less capa-
the question whether maintaining cell homeosta- ble of responding to a chemotactic stimulus and
sis by growth factors, such as VEGF, could are less potent in contracting three-dimensional
rescue emphysema. Unfortunately, no data are collagen gels [Rennard et al. 2006].
so far available on the potential benefits of
VEGF gene therapy in emphysematous lung. It is unclear whether the differences between
VEGF is a tightly regulated gene in the lung, fibroblasts obtained from normal or emphysema-
playing specific roles in different structural and tous individuals represent underlying genetic dif-
cellular compartments. While the VEGF protein ferences and hence susceptibility to develop
and mRNA contents in the lung are reduced in emphysema, or if they are an acquired defect.
severe emphysema, VEGF gene expression is
increased in the pulmonary arteries of smokers Bone marrow-derived cells
and patients with moderate COPD, and corre- Circulating bone marrow-derived progenitors
lates with medial thickening of the pulmonary have been shown to be decreased in COPD
vascular walls [Santos et al. 2003]. These data patients and to correlate with disease severity
confirm the systemic pathogenesis of emphy- [Huertas and Palange, 2011; Huertas et al.
sema, but also underlie the multifactorial aspect 2010; Palange et al. 2006; Fadini et al. 2006].
of the disease [Yoshida and Tuder, 2007]. Several clinical factors have been implicated in
the mobilization of endothelial progenitor cells
Repair failure (EPCs), and mechanisms have begun to be elu-
The destruction of the alveolar walls leading to cidated. Defective lung development or defective
enlargement of the alveolar spaces, which is a lung repair in the setting of protracted inflamma-
well known characteristic of emphysema, could tion and injury may result in part from an inad-
also be considered as the result of an impaired equate contribution of local or circulating EPCs.
repair capacity. In addition to this, there is an Age has previously been reported to be inversely
indication that lung repair, as evidenced by de correlated with EPC number [Chang et al. 2007].
novo synthesis and tissue accumulation of elastin Newer data suggest that there are also differences
and collagen, is inhibited by cigarette smoke in the ability of EPCs to home to ischemic tissues
[Rennard et al. 2006]. Exposure to cigarette based on age, and that this may be mediated
smoke extract also inhibits fibroblast proliferation through the inability of aged tissues to activate
[Nakamura et al. 1995], and fibroblasts isolated normally the hypoxia-inducible factor-
from patients with emphysema exhibit decreased 1a-mediated hypoxia response [Llevadot et al.
proliferative capacity [Holz et al. 2004; Nobukuni 2001].
et al. 2002]. Exposure to cigarette smoke extract
induces cell-cycle arrest in fibroblasts, mediated It has also been shown that in COPD patients,
through the activation of p53 and p16, which bone marrow dysfunction is related to lung func-
inhibit the cell cycle, leading to cellular senes- tion impairment, poor nutritional state, and
cence [Nyunoya et al. 2006]. This may represent levels of systemic inflammation [Huertas et al.
a response of fibroblasts to DNA damage by cig- 2010]. In this study, patients with low BMI had
arette smoke extract [Bartek et al. 2004]. This reduced circulating hemopoietic and endothelial
may result in abnormal wound healing and progenitor counts, suggesting lower progenitor

http://tar.sagepub.com 219

Downloaded from tar.sagepub.com at UNIV OF LETHBRIDGE on August 19, 2014


Therapeutic Advances in Respiratory Disease 5 (3)

release from the bone marrow, pronounced sys- [Sato et al. 2006]. Interestingly, in experimental
temic inflammation with high levels of proangio- models of emphysema, genes that encode proteins
genetic growth factors, and proinflammatory important for the systemic immune response have
markers, compared with normal BMI patients. been shown to be involved. Mice lacking toll-like
Interestingly, these growth factors and cytokines, receptor 4, which is activated during the innate
as well as BMI, were inversely correlated with immune response, spontaneously developed
markers of disease severity. Furthermore, emphysema associated with an oxidative stress
among patients with similar pulmonary impair- imbalance (i.e. increased Nox3 gene expression,
ment, those who displayed low BMI had a greater a novel NAPDH oxidase, and elastin degradation)
bone marrow dysfunction. [Zhang et al. 2006], but without any inflammatory
infiltration of the lung parenchyma.
All together, these data suggest that, in addition
to disease progression in terms of airflow obstruc- Lung gene expression studies from endstage
tion, systemic involvement in COPD leads to a COPD patients have shown impaired mitochon-
more pronounced bone marrow impairment. drial energy metabolism and protein synthesis
Whether this pool of precursor cells is reduced [Golpon et al. 2004], suggesting DNA damage
as a consequence of the pulmonary disease or and posttranscriptional modifications, but little
whether the bone marrow involvement represents is known about epigenetic marks in respiratory
a systemic disease needs to be defined. diseases. Interestingly, it has been shown in
COPD that the expression and activity of histone
Genetics and epigenetics deacetylase (HDAC)-2 are markedly reduced in
The genetic etiology of COPD is certainly in favor lung parenchyma, bronchial biopsies, and alveo-
of considering the disease as a systemic syndrome: lar macrophages. This decrease is correlated with
although smoking is considered to be the major disease severity and intensity of the inflammatory
risk factor for COPD, only 15% to 20% of smo- response [Ito et al. 2005]. Overexpression or
kers develop the clinically relevant disease [Celli knockdown of HDAC2 in alveolar macrophages
and MacNee, ATS/ERS Task Force, 2004; from COPD patients affects not only the inflam-
Ouellette, 2004]. Genetic susceptibility to matory response but also corticosteroid respon-
COPD has been investigated in recent years and siveness, suggesting an additional role for
numerous candidate genes have been found to HDAC2 in the anti-inflammatory actions of the
encode proteins that regulate: proteases and anti- corticosteroid [Ito et al. 2006].
proteases (i.e. 1-antitrypsin, Serpine2, 1-antichy-
motrypsin, 2-macroglobulin, secretory leukocyte The clinical impact of cigarette smoking, which
proteinase inhibitor, matrix metalloproteinases ranges from negligible to endstage lung disease,
(MMP), ADAM33, protease-activated receptor-2); is, in part, determined by individual susceptibility
mucociliary clearance (i.e. cystic fibrosis transmem- factors. Understanding the factors that lead to
brane regulator, mucins); antioxidants (i.e. micro- emphysema is useful for predicting high-risk
somal epoxide hydrolase, glutathione-S-transferases, individuals. Except for a1-antitrypsin, a defi-
cytochrome P450, extracellular superoxide dismut- ciency of which results in a predisposition to
ase); inflammatory mediators (i.e. TNF- , IL-11, smoking-induced emphysema, few other suscep-
IL-1 family). Interestingly, the MMP-1 promoter tibility factors are defined. Genetic susceptibility
has been shown to be a direct target of cigarette and epigenetic modifications strongly suggest a
smoke in lung epithelial cells and regions of the systemic etiology of COPD, but further data are
human MMP-1 promoter have been recently needed to define better the precise etiopathology
defined [Mercer et al. 2009]. This extensive list of the disease.
of candidate genes attests to the relevance of the
genetic pathophysiology of COPD and, in partic- Immunology
ular, emphysema. Many animal models have An interesting characteristic of emphysema is the
helped in defining the genes involved in the path- lack of attenuation in inflammatory and disease
ogenesis of this latter disorder, strongly suggesting parameters years after the cessation of smoking:
the systemic origin of the disease. Recently, cellu- tobacco smoke seems to contain antigens that
lar senescence has also been shown in a mouse induce immunological responses in susceptible
model of emphysema, where knocking out the individuals. It might also be a more complex
senescence marker protein-30, which protects pathogenesis in which smoking induces an auto-
against aging, leads to airspace enlargement immune response to an endogenous lung antigen.

220 http://tar.sagepub.com

Downloaded from tar.sagepub.com at UNIV OF LETHBRIDGE on August 19, 2014


A Huertas and P Palange

This alternative concept of emphysema patho- Infections


genesis has been raised by Voelkel and colleagues, Other observations keep the question of COPD
who showed that humoral and CD4þ cell- as a systemic disease open: emphysematous lung
dependent mechanisms are sufficient to trigger destruction has been reported in other nonsmok-
emphysema development, suggesting that alveo- ing-related disorders or hypersensitivity pneumo-
lar septal cell destruction might result from nitis [Tuder and Voelkel, 2001], and recent data
immune mechanisms [Taraseviciene-Stewart indicate that starvation causes lung cell apoptosis
et al. 2005]. Using a murine model, they in mouse lung [Massaro et al. 2004].
showed that intraperitoneal injection of endothe- Furthermore, in a recent observational study,
lial cells causes emphysema, leading to alveolar HIV was identified as an independent risk
septal cell apoptosis, and the activation of factor for COPD after adjusting for age, race,
MMP-9 and MMP-2. Furthermore, naı̈ve pack-years of smoking, intravenous drug abuse,
immunocompetent animals developed emphy- and alcohol abuse. HIV subjects were 50—60%
sema following the administration of CD4þ T more likely to receive a diagnosis of COPD
cells. This hypothesis has also been investigated when these risk factors were taken into accounted
in humans: by isolating peripheral blood CD4þ [Crothers et al. 2011, 2006]. A series of 114
T cells from emphysematous patients, Lee and HIVþ patients, when compared with 44 HIV—
colleagues were able to show that exposure to cig- controls, revealed a significantly higher rate of
arette smoke induces secretion of proteolytic emphysema among the HIVþ individuals [Diaz
enzymes from cells of the innate immune et al. 2000]. When controlled for tobacco expo-
system. These cells liberate lung elastin fragments sure, the HIVþ subjects again had significantly
which, in susceptible individuals, could initiate T more emphysematous damage for a given level
cell and B cell-mediated immunity against elastin of tobacco use. HIV is now frequently cited as a
[Lee et al. 2007]. These data strongly suggest an susceptibility factor for the development of
autoimmune pathogenesis of emphysema, charac- emphysema, independently of cigarette smoking
terized by the presence of antielastin antibody and status. The presence of common coexistent fac-
T helper type 1 (Th1) responses that correlate tors that may predispose patients to chronic lung
with emphysema severity. injury such as drugs, opportunistic infections,
and malnutrition, limits the scope of studies of
It is interesting to point out that elastin is abun- direct mechanisms involved in HIV-associated
dant in various tissues, in particular in arteries, emphysematous lung disease. Adding to the com-
arterioles, and skin. In addition to emphysema, plexity of HIV-associated COPD, several risk fac-
tobacco smokers are at high risk for coronary tors associated with HIV infection may
artery disease, aortic aneurysms, and elastolytic themselves play a pathogenic role in the develop-
changes of the skin [Patel et al. 2006; Pepine et al. ment of emphysema, such as infection or coloni-
2006]. These findings link emphysema to adap- zation with Pneumocystis, intravenous drug use,
tive immunity against a specific lung antigen and malnutrition, etc. In the non-HIV infected pop-
suggest the risk for smokers of developing auto- ulation, Pneumocystis colonization has been asso-
immune diseases in other elastin-rich tissues, ciated with increased severity of airway
such as arteries and skin. Findings of antielastin obstruction in COPD [Morris et al. 2004;
autoimmunity in emphysema therefore suggest a Palange et al. 1994]. In the studied cohort of
broader, systemic autoimmune process involving patients, 36.7% of patients with GOLD stage
the major elastin-bearing organs that may explain IV were colonized with Pneumocystis compared
these diverse clinical observations. with only 5.3% of patients with less severe
COPD or normal lung function. These data led
On the other hand, several clinical reports have to the hypothesis that Pneumocystis colonization
described cases of emphysema in patients with may accelerate the development of airway
hypocomplementemia, and in particular, those obstruction. Further studies of the pathogenic
with a history of hypocomplementemic urticarial role of fungal colonization in the development
vasculitis. Although we need to better define this of airspace enlargement in emphysema are
possible pathogenesis, these reports suggest that needed to understand better the role of coloniza-
an impaired immunological condition could lead tion by Pneumocystis and infection in the patho-
to the development of emphysema [Jamison et al. genesis of HIV-related emphysema. HIV-
2008; Ghamra and Stoller, 2003], underlying the seropositive individuals have a propensity to
systemic pattern of the disease. develop emphysematous changes in their upper

http://tar.sagepub.com 221

Downloaded from tar.sagepub.com at UNIV OF LETHBRIDGE on August 19, 2014


Therapeutic Advances in Respiratory Disease 5 (3)

lobes at an accelerated rate independently of their Barnes, P.J. and Celli, B.R. (2009) Systemic manifes-
smoking status. Confounding factors in HIV- tations and comorbidities of COPD. Eur Respir J
33: 1165—1185.
infected individuals, including Pneumocystis colo-
nization, drug use, and malnutrition, may play a Bartek, J., Lukas, C. and Lukas, J. (2004) Checking on
role in predisposition for emphysema. However, DNA damage in S phase. Nat Rev Mol Cell Biol
5: 792—804.
HIV itself is emerging as an independent factor in
the pathogenesis of emphysema. A recent review Biskobing, D.M. (2002) COPD and osteoporosis.
[Petrache et al. 2008] emphasized how HIV Chest 121: 609—620.
infection may affect cytotoxic lymphocyte activa- Bolton, C.E., Ionescu, A.A., Shiels, K.M., Pettit, R.J.,
tion, lung capillary endothelial cell injury and Edwards, P.H., Stone, M.D. et al. (2004) Associated
apoptosis, sphingolipid imbalance, and oxidative loss of fat-free mass and bone mineral density in
stress in the lung. A better understanding of the chronic obstructive pulmonary disease. Am J Respir
Crit Care Med 170: 1286—1293.
pathogenesis of HIV-associated pulmonary
emphysema may provide clues and therapeutic Carmeliet, P., Ferreira, V., Breier, G., Pollefeyt, S.,
targets that have broader application in this dis- Kieckens, L., Gertsenstein, M. et al. (1996) Abnormal
blood vessel development and lethality in embryos
ease, including cigarette smoke-induced
lacking a single VEGF allele. Nature 380: 435—439.
emphysema.
Celli, B.R. and MacNee, W. for the ATS/ERS Task
Force (2004) Standards for the diagnosis and treat-
Conclusion
ment of patients with COPD: A summary of the ATS/
Many extrapulmonary effects of COPD have ERS position paper. Eur Respir J 23: 932—946.
been described in the last two decades: some of
these manifestations have been extensively stud- Chang, E.I., Loh, S.A., Ceradini, D.J., Chang, E.I.,
Lin, S.E., Bastidas, N. et al. (2007) Age decreases
ied, but it is still unclear whether they represent endothelial progenitor cell recruitment through
consequences of the pulmonary disorder, or decreases in hypoxia-inducible factor 1alpha stabiliza-
whether COPD should be considered as a sys- tion during ischemia. Circulation 116: 2818—2829.
temic disease. In this review, we have tried to Creutzberg, E.C., Schols, A.M., Weling-Scheepers,
give an overview of the most relevant and inno- C.A., Buurman, W.A. and Wouters, E.F.M. (2000)
vative hypothesis about the extrapulmonary Characterization of nonresponse to high caloric oral
effects of COPD. Even if precise cellular and nutritional therapy in depleted patients with chronic
molecular mechanisms are still unclear, evidence obstructive pulmonary disease. Am J Respir Crit Care
Med 161: 745—752.
has been provided suggesting that COPD, and
emphysema in particular, might be considered Crothers, K., Butt, A.A., Gibert, C.L., Rodriguez-
as a systemic disease. Barradas, M.C., Crystal, S. and Justice, A.C. for the
Veterans Aging Cohort 5 Project Team. (2006)
Increased COPD among HIV-positive compared to
Further studies are needed to understand better HIV-negative veterans. Chest 130: 1326—1333.
this complex and multifaceted disease in order to
treat patients with more specific tools. Crothers, K., Huang, L., Goulet, J.L., Goetz, M.B.,
Brown, S., Rodriguez-Barradas, M. et al. (2011)
HIV infection and risk for incident
Funding pulmonary diseases in the combination antiretroviral
This research received no specific grant from any therapy era. Am J Respir Crit Care Med 183: 388—395.
funding agency in the public, commercial, or not- Diaz, P.T., King, M.A., Pacht, E.R., Wewers, M.D.,
for-profit sectors. Gadek, J.E., Nagaraja, H.N. et al. (2000) Increased
susceptibility to pulmonary emphysema among HIV-
Conflict of interest statement seropositive smokers. Ann Intern Med 132: 369—372.
The authors declare that there are no conflicts of Fadini, G.P., Schiavon, M., Cantini, M., Baesso, I.,
interest. Facco, M., Miorin, M. et al. (2006) Circulating pro-
genitor cells are reduced in patients with severe lung
disease. Stem Cells 24: 1806—1810.
References Gan, W.Q., Man, S.F., Senthilselvan, A. and Sin, D.D.
Agustı́, A.G., Noguera, A., Sauleda, J., Sala, E., Pons, (2004) Association between chronic obstructive pul-
J. and Busquets, X. (2003) Systemic effects of chronic monary disease and systemic inflammation: A sys-
obstructive pulmonary disease. Eur Respir J tematic review and a meta-analysis. Thorax
21: 347—360. 59: 574—580.
Agusti, A.G. and Soriano, J.B. (2008) COPD as a Ghamra, Z. and Stoller, J.K. (2003) Basilar hyperlu-
systemic disease. COPD 5: 133—138. cency in a patient with emphysema due to

222 http://tar.sagepub.com

Downloaded from tar.sagepub.com at UNIV OF LETHBRIDGE on August 19, 2014


A Huertas and P Palange

hypocomplementemic urticarial vasculitis syndrome. Lee, S.H., Goswami, S., Grudo, A., Song, L.Z.,
Respir Care 48: 697—699. Bandi, V., Goodnight-White, S. et al. (2007)
Antielastin autoimmunity in tobacco smoking-induced
Golpon, H.A., Coldren, C.D., Zamora, M.R., emphysema. Nat Med 13: 567—569.
Cosgrove, G.P., Moore, M.D., Tuder, R.M. et al.
(2004) Emphysema lung tissue gene expression pro- Liebow, A.A. (1959) Pulmonary emphysema with
filing. Am J Respir Cell Mol Biol 31: 595—600. special reference to vascular changes. Am Rev Respir
Dis 80: 67—93.
Gray-Donald, K., Gibbons, L., Shapiro, S.H.,
Macklem, P.T. and Martin, J.G. (1996) Nutritional Llevadot, J., Murasawa, S., Kureishi, Y., Uchida, S.,
status and mortality in chronic obstructive pulmonary Masud, H., Kawamoto, A. et al. (2001) HMG-CoA
disease. Am J Respir Crit Care Med 153: 961—966. reductase inhibitor mobilizes bone marrow-derived
endothelial progenitor cells. J Clin Invest
Holz, O., Zuhlke, I., Jaksztat, E., Muller, K.C., 108: 399—405.
Welker, L., Nakashima, M. et al. (2004) Lung fibro-
blasts from patients with emphysema show a reduced Massaro, D., Massaro, G.D., Baras, A., Hoffman, E.P.
proliferation rate in culture. Eur Respir J 24: 575—579. and Clerch, L.B. (2004) Calorie-related rapid onset of
alveolar loss, regeneration, and changes in mouse lung
Hoshino, S., Yoshida, M., Inoue, K., Yano, Y., gene expression. Am J Physiol Lung Cell Mol Physiol
Yanagita, M., Mawatari, H. et al. (2005) Cigarette 286: L896—L906.
smoke extract induces endothelial cell injury via JNK
pathway. Biochem Biophys Res Commun 329: 58—63. McGrath-Morrow, S.A., Cho, C., Zhen, L., Hicklin,
D.J. and Tuder, R.M. (2005) Vascular endothelial
Huertas, A. and Palange, P. (2011) Circulating pro- growth factor receptor 2 blockade disrupts postnatal
genitor cells and chronic pulmonary diseases. Eur lung development. Am J Respir Cell Mol Biol
Respir J 37: 426—431. 32: 420—427.
Huertas, A., Testa, U., Riccioni, R., Petrucci, E., Riti, Mercer, B.A., Wallace, A.M., Brinckerhoff, C.E. and
V., Savi, D. et al. (2010) Bone marrow-derived pro- D’Armiento, J.M. (2009) Identification of a cigarette
genitors are greatly reduced in patients with severe smoke responsive region in the distal MMP-1 pro-
COPD and low-BMI. Respir Physiol Neurobiol moter. Am J Respir Cell Mol Biol 40: 4—12.
170: 23—31.
Morris, A., Sciurba, F.C., Lebedeva, I.P., Githaiga, A.,
Hurst, J.R., Wilkinson, T.M., Perera, W.R., Elliott, W.M., Hogg, J.C. et al. (2004) Association of
Donaldson, G.C. and Wedzicha, J.A. (2005) chronic obstructive pulmonary disease severity and
Relationships among bacteria, upper airway, lower Pneumocystis colonization. Am J Respir Crit Care Med
airway, and systemic inflammation in COPD. Chest 170: 408—413.
127: 1219—1226.
Nakamura, Y., Romberger, D.J., Tate, L., Ertl, R.F.,
Ito, K., Ito, M., Elliott, W.M., Cosio, B., Caramori, Kawamoto, M., Adachi, Y. et al. (1995) Cigarette
G., Kon, O.M. et al. (2005) Decreased histone dea- smoke inhibits lung fibroblast proliferation and che-
cetylase activity in chronic obstructive pulmonary dis- motaxis. Am J Respir Crit Care Med 151: 1497—1503.
ease. N Engl J Med 352: 1967—1976.
Nobukuni, S., Watanabe, K., Inoue, J., Wen, F.Q.,
Ito, K., Yamamura, S., Essilfie-Quaye, S., Cosio, B., Tamaru, N. and Yoshida, M. (2002) Cigarette smoke
Ito, M., Barnes, P.J. et al. (2006) Histone deacetylase inhibits the growth of lung fibroblasts from patients
2-mediated deacetylation of the glucocorticoid with pulmonary emphysema. Respirology 7: 217—223.
receptor enables NF-kB suppression. J Exp Med
203: 7—13. Nyunoya, T., Monick, M.M., Klingelhutz, A.,
Yarovinsky, T.O., Cagley, J.R. and Hunninghake, G.W.
John, M., Hoernig, S., Doehner, W., Okonko, D.D., (2006) Cigarette smoke induces cellular senescence.
Witt, C. and Anker, S.D. (2005) Anemia and inflam- Am J Respir Cell Mol Biol 35: 681—688.
mation in COPD. Chest 127: 825—829.
Ouellette, D.R. (2004) The answer is fifteen percent:
Jamison, S.C., Brierre, S., Sweet, J. and de Boisblanc, What is the question? Chest 125: 3—5.
B. (2008) A case of precocious emphysema and lung
cancer in a woman with a history of hypocomplemen- Palange, P. (1998) Renal and hormonal abnormalities
temic urticarial vasculitis. Chest 133: 787—789. in chronic obstructive pulmonary disease (COPD).
Thorax 53: 89—91.
Kasahara, Y., Tuder, R.M., Taraseviciene-Stewart,
L., Le Cras, T.D., Abman, S., Hirth, P.K. et al. Palange, P., Serra, P., Di Sorbato, F., Contini, C. and
(2000) Inhibition of VEGF receptors causes lung cell Giacovazzo, M. (1994) Pneumocystis carinii pneumonia
apoptosis and emphysema. J Clin Invest in a patient with chronic obstructive pulmonary dis-
106: 1311—1319. ease but no evident immunoincompetence. Clin Infect
Dis 19: 543—544.
Langen, R.C., Schols, A.M., Kelders, M.C., Wouters,
E.F. and Janssen-Heininger, Y.M. (2001) Palange, P., Testa, U., Huertas, A., Calabrò, L.,
Inflammatory cytokines inhibit myogenic differentia- Antonucci, R., Petrucci, E. et al. (2006) Circulating
tion through activation of nuclear factor-kappaB. haemopoietic and endothelial progenitor cells are
FASEB J 15: 1169—1180. decreased in COPD. Eur Respir J 27: 529—541.

http://tar.sagepub.com 223

Downloaded from tar.sagepub.com at UNIV OF LETHBRIDGE on August 19, 2014


Therapeutic Advances in Respiratory Disease 5 (3)

Patel, B.D., Loo, W.J., Tasker, A.D., Screaton, N.J., systemic inflammation in chronic obstructive pulmo-
Burrows, N.P., Silverman, E.K. et al. (2006) Smoking nary disease. Circulation 107: 1514—1519.
related COPD and facial wrinkling: Is there a common
susceptibility? Thorax 61: 568—671. Steiner, M.C., Barton, R.L., Singh, S.J. and Morgan,
M.D.L. (2003) Nutritional enhancement of exercise
Pepine, C.J., Kowey, P.R., Kupfer, S., Kolloch, R.E., performance in chronic obstructive pulmonary disease:
Benetos, A., Mancia, G. et al. (2006) Predictors of A randomised controlled trial. Thorax 58: 745—751.
adverse outcome among patients with hypertension
Tang, K., Rossiter, H.B., Wagner, P.D. and Breen,
and coronary artery disease. J Am Coll Cardiol
E.C. (2004) Lung-targeted VEGF inactivation leads to
47: 547—551.
an emphysema phenotype in mice. J Appl Physiol
Petrache, I., Diab, K., Knox, K.S., Twigg, H.L., 97: 1559—1566.
Stephens, R.S., Flores, S. et al. (2008) HIV associated
Taraseviciene-Stewart, L., Scerbavicius, R., Choe,
pulmonary emphysema: A review of the literature and K.H., Moore, M., Sullivan, A., Nicolls, M.R. et al.
inquiry into its mechanism. Thorax 63: 463—469. (2005) An animal model of autoimmune emphysema.
Remels, A.H., Gosker, H.R., van der Velden, J., Am J Respir Crit Care Med 171: 734—742.
Langen, R.C. and Schols, A.M. (2007) Systemic Taraseviciene-Stewart, L. and Voelkel, N.F. (2008)
inflammation and skeletal muscle dysfunction in Molecular pathogenesis of emphysema. J Clin Invest
chronic obstructive pulmonary disease: State of the art 118: 394—402.
and novel insights in regulation of muscle plasticity.
Clin Chest Med 28: 537—552. Tuder, R.M. and Voelkel, N.F. (2001) Pathology of
chronic bronchitis and emphysema, In: Voelkel, N.F.
Rennard, S.I., Togo, S. and Holz, O. (2006) Cigarette and MacNee, W. (eds). Chronic Obstructive Lung
smoke inhibits alveolar repair: A mechanism for the Disease, Dekker: Montreal.
development of emphysema. Proc Am Thorac Soc
3: 703—708. Vernooy, J.H., Küçükaycan, M., Jacobs, J.A.,
Chavannes, N.H., Buurman, W.A., Dentener, M.A.
Santos, S., Peinado, V.I., Ramı́rez, J., Melgosa, T., et al. (2002) Local and systemic inflammation in
Roca, J., Rodriguez-Roisin, R. et al. (2002) patients with chronic obstructive pulmonary disease:
Characterization of pulmonary vascular remodelling in Soluble tumor necrosis factor receptors are increased
smokers and patients with mild COPD. Eur Respir J in sputum. Am J Respir Crit Care Med
19: 632—638. 166: 1218—1224.
Santos, S., Peinado, V.I., Ramirez, J., Morales-Blanhir, Voelkel, N.F. and Taraseviciene-Stewart, L. (2005)
J., Bastos, R., Roca, J. et al. (2003) Enhanced expres- Emphysema: An autoimmune vascular disease? Proc
sion of vascular endothelial growth factor in pulmo- Am Thorac Soc 2: 23—25.
nary arteries of smokers and patients with moderate
chronic obstructive pulmonary disease. Am J Respir Wang, H., Liu, X., Umino, T., Sköld, C.M., Zhu, Y.,
Crit Care Med 167: 1250—1256. Kohyama, T. et al. (2001) Cigarette smoke inhibits
human bronchial epithelial cell repair processes. Am J
Sato, T., Seyama, K., Sato, Y., Mori, H., Souma, S., Respir Cell Mol Biol 25: 772—779.
Akiyoshi, T. et al. (2006) Senescence marker protein-
30 protects mice lungs from oxidative stress, aging, Wouters, E.F. (2002) Systemic effects in COPD. Chest
and smoking. Am J Respir Crit Care Med 121: 127S—130S.
174: 530—537. Yamato, H., Sun, J.P., Churg, A. and Wright, J.L.
Schols, A.M., Slangen, J., Volovics, L. and Wouters, (1996) Cigarette smoke-induced emphysema in guinea
E.F.M. (1998) Weight loss is a reversible factor in the pigs is associated with diffusely decreased capillary
density and capillary narrowing. Lab Invest
prognosis of chronic obstructive pulmonary disease.
75: 211—219.
Am J Respir Crit Care Med 157: 1791—1797.
Yoshida, T. and Tuder, R.M. (2007) Pathobiology of
Shapiro, S.D. (2000) Vascular atrophy and VEGFR-2
cigarette smoke-induced chronic obstructive pulmo-
signaling: Old theories of pulmonary emphysema meet
nary disease. Physiol Rev 87: 1047—1082.
new data. J Clin Invest 106: 1309—1310.
Visit SAGE journals online Zhang, X., Shan, P., Jiang, G., Cohn, L. and Lee, P.J.
http://tar.sagepub.com Sin, D.D. and Man, S.F. (2003) Why are patients with
(2006) Toll-like receptor 4 deficiency causes pulmo-
chronic obstructive pulmonary disease at increased nary emphysema. J Clin Invest 116: 3050—3059.
risk of cardiovascular diseases? The potential role of

224 http://tar.sagepub.com

Downloaded from tar.sagepub.com at UNIV OF LETHBRIDGE on August 19, 2014

You might also like