• Nie Znaleziono Wyników

Pathogenesis of chronic obstructive pulmonary disease

N/A
N/A
Protected

Academic year: 2022

Share "Pathogenesis of chronic obstructive pulmonary disease"

Copied!
7
0
0

Pełen tekst

(1)

Adres do korespondencji:

Adres do korespondencji:

Adres do korespondencji:

Adres do korespondencji:

Adres do korespondencji: prof. Steven I. Renard M.D., University of Nebrasca Medical Center, Omaha, NE, e-mail: srennard@unma.edu Praca wpłynęła do Redakcji: 6.12.2010 r.

Copyright © 2011 Via Medica ISSN 0867–7077

Stephen I. Rennard

University of Nebraska Medical Center, Omaha, NE

Pathogenesis of chronic obstructive pulmonary disease

Patogeneza przewlekłej obturacyjnej choroby płuc

Abstract

Current concepts suggest that COPD largely results from tissue injury, most commonly consequent to an exposure, the most important of which is cigarette smoke. This injury is mediated by a complex network of inflammatory cells and mediators, and it is likely that heterogeneity in the inflammatory response accounts for some of the variable susceptibility to develop COPD and for the some of the clinical heterogeneity of the disease. Mechanisms that can prevent tissue injury are also diverse and are inhibited to varying degrees among individuals with COPD. Finally, the alterations in tissue structure that compromise function depend not only on tissue injury, but also on tissue repair, which can either mitigate or exacerbate lung function loss depending on whether repair processes are effective or disruptive.

Pneumonol. Alergol. Pol. 2011; 79, 2: 132–138

Streszczenie

Współczesne poglądy na patogenezę POChP sugerują, że choroba jest spowodowana uszkodzeniem płuc najczęściej spowodowanym przez dym tytoniowy. Uszkodzenie płuc następuje w wyniku złożonego współdziałania komórek zapalnych i mediatorów zapalenia. Jest prawdopodobne, że różnorodność odpowiedzi zapalnej na dym tytoniowy jest odpowiedzialna za zmienną podatność na rozwój choroby a także różnorodność obrazu klinicznego. Mechanizmy chroniące płuca przed uszkodzeniem są też różnorodne i są w różnym stopniu zahamowane u indywidualnych chorych. Ostatecznie, anatomiczne uszkodzenie płuc, które upośledza czynność płuc zależy nie tylko od procesów niszczenia lecz także od procesów naprawczych w płucach, które mogą chronić czynność płuc lub nasilać zaburzenia, w zależności czy procesy naprawcze są skuteczne czy nasilające uszkodzenie.

Pneumonol. Alergol. Pol. 2011; 79, 2: 132–138

Introduction

Chronic obstructive pulmonary disease (COPD) is a major cause of morbidity and mortali- ty worldwide [1]. The most important cause is ci- garette smoking, and studies over the last sixty years have outlined the mechanisms by which smoke exposure can lead to disease [2]. COPD is also recognized as being markedly heterogeneous [3, 4], and it is likely that this is due to multiple pathogenetic mechanisms having different roles in individual patients [2]. It is hoped that understan- ding the pathogenesis of COPD will lead to novel

therapies and that understanding the mechanisms important in individual patients will allow these therapies to be used selectively [3, 4]. This review will provide an overview of COPD pathogenesis with some emphasis on the newer concepts that altered tissue repair can play a key role.

Historical concepts

Historical concepts of COPD pathogenesis were centered on physiological mechanisms.

COPD is defined by airflow limitation, but this can result from disease of either the airways or the

(2)

macrophages that are resident in the lung, smoke can initiate the release of a variety of chemokines.

These, in turn, can mediate the recruitment and activation of inflammatory cells. Lung cells can also release growth factors that act on bone mar- row to increase the production of inflammatory cells. A number of factors capable of mediating the recruitment of neutrophils, monocyte/macropha- ges and lymphocytes have all been reported in this regard. In addition, mediators capable of recruiting inflammatory cells can also be generated from de- gradation products of extracellular matrix. This suggests that inflammation, by inducing tissue da- mage, could initiate a persistent “vicious circle.”

The relative importance of the various factors that can recruit inflammatory cells is not establi- shed. Interleukin (IL)-8, which is induced by smo- ke and increased in the lungs of smokers and COPD patients, has attracted considerable attention [2, 11, 12]. A pilot clinical trial with an anti-IL-8 anti- body, however, improved dyspnea, but did not al- ter lung function. The lack of a dramatic effect supports the concept that multiple mediators may modulate inflammatory cell recruitment and im- plies that therapies targeting a specific mediator may not be effective, although this trial did not directly assess inflammation.

In addition to activating cellular inflamma- tory responses, cigarette smoke can also activate humoral inflammation mediated by complement.

Smoke activates complement [13] and leads to the generation of C5a, a potent chemotactic factor [14]. The activity of C5a is greatly potentiated by a co-factor, Gc-globlulin, also known as vitamin D binding protein [15]. This, in turn, is blocked by the protein chemotactic factor inhibitor (CFI).

Smoke can inhibit the activity of CFI, thus leading to unopposed Gc-globulin potentiated C5a chemo- taxis [16].

Inflammatory mediators and tissue damage The concept that neutrophil elastase media- tes tissue damage has been greatly expanded [2].

The neutrophil produces two serine proteases in addition to elastase, and all three can cause em- physema in model systems. In addition, neutro- phils produce several mediators in the other ma- jor classes: matrix metalloproteases (MMPs) and cystein proteases, and these can also result in tis- sue damage and emphysema. Other inflammatory cells produce their own sets of secreted proteases and, as a result, there is a large number of prote- ases likely present in the lower respiratory tract of smokers and COPD patients that can cause tissue damage. In addition, proteases likely contribute to pulmonary parenchyma [5]. The degree to which

these different sites are involved can vary marke- dly from patient to patient. Disease of the large airways results in chronic cough with sputum pro- duction and disease of the small airways causes airflow limitation [2]. Disease of the pulmonary pa- renchyma that results in loss of alveolar tissue with enlargement of airspaces, i.e. emphysema, may or may not be associated with airflow limitation [5].

Orie and colleagues from the Netherlands pro- posed the idea that asthma, through the chronic effects of airways hyper-reactivity, could lead to fixed narrowing of the airways and airflow obstruc- tion [6]. This concept was contrasted with the idea that airways infection leads to mucus hypersecre- tion, which, over time, leads to airflow obstruction [7]. Both concepts have become more complex over time, and current data suggests that both “path- ways” contribute to airflow limitation [2].

Laurell and Erickson described that emphyse- ma occurs among individuals deficient in the cir- culating serine protease inhibitor alpha 1 anti-tryp- sin [8]. This led to the concept that imbalance be- tween protease production and anti-protease pro- tection leads to tissue destruction. About the same time, Liebow suggested that failure to maintain an adequate pulmonary capillary bed should lead to loss of alveolar wall and the development of em- physema [9]. A large body of data now supports the general concept of an imbalance between me- diators that damage tissue and defense mechani- sms that provide protection, and a growing body of evidence supports the concept that failure of tis- sue maintenance and repair also contribute to emphysema.

Importantly, current concepts, which are ba- sed on the cellular and biochemical pathways in- volved, suggest that shared mechanisms contri- bute to disease pathogenesis at different sites and help explain the overlap among the historical hy- potheses.

Cigarette smoke and inflammation Cellular recruitment and activation

The concept that elastase released from neu- trophils exceeded the protective effect of alpha 1 anti-trypsin and resulted in tissue damage led to many studies of inflammation and inflammatory mediators in the lungs of COPD patients and in response to cigarette smoke. Smoke, which conta- ins as many as 6,000 molecular entities, can ini- tiate an inflammatory response in a number of ways [2, 10]. By interacting with the epithelial cells that line the airways and alveoli as well as the

(3)

COPD pathogenesis by activating cells. Neutrophil elastase, for example, is an extremely potent sti- mulus for mucus release and induces goblet cell metaplasia in the airways [17–19]. Thus, proteases may play a pathogenetic role in airways disease as well as emphysema.

In addition to the release of proteases, inflam- matory cells can induce tissue damage through the release of both active oxidants and toxic peptides [2]. The oxidative stress induced by smoke, in par- ticular, has been shown in a number of model sys- tems to lead to changes that resemble COPD. In addition, the lower respiratory tract of patients with COPD shows signs of oxidative stress [20].

Treatment with anti-oxidants, however, has had limited success [21]. As with proteases, the lung contains a number of endogenous anti-oxidants.

How the balance among the various oxidant and anti-oxidant species present in the lung contribu- tes to the pathogenesis of COPD is an area of acti- ve research.

Inflammatory cells also produce peptides that can damage lung cells. These include peptides that have non-specific toxicity such as defensins and granzyme B [22] as well as small molecules such as ceramide [23] that can induce cellular apopto- sis. By inducing cell damage, these mediators may lead to the development of emphysema.

In addition to stimulating the recruitment and activation of inflammatory cells, smoke can also shift the protease burden of the lung by inactiva- ting anti-proteases and anti-oxidants. Alpha 1 anti- trypsin, which also inhibits the activity of defen- sins, for example, is inactivated by cigarette smo- ke [2, 24]. Neutrophil elastase can degrade TIMP- 1, a major inhibitor of MMPs [2]. Thus smoke, by inactivation of alpha 1 anti-trypsin, could cause excess elastase burden that, in turn, will reduce levels of MMP1, which will result in less inhibi- tion of MMP activity. Because of the multiple in- teractions among the various protease cascades, cigarette smoke can lead to a variety of proteolytic activities.

Other inflammatory etiologies of COPD Cigarette smoking is the most important cau- sative factor for COPD. However, many factors in addition to smoke can also contribute. Other expo- sures such as environmental or indoor air pollu- tion or a work enviroment with dusts or fumes have been well documented to contribute to COPD [10].

These exposures likely activate similar pathogene- tic mechanisms as does cigarette smoke.

It is likely that other risk factors also lead to excess lung inflammation. Asthma, for example,

can progress to fixed airflow limitation even in the absence of smoking [25]. It has been suggested that airway remodeling consequent to the airway in- flammation that characterizes asthma is the prime mechanism for this process.

Persistence of inflammation

A major feature of COPD is that, once establi- shed, the disease progresses despite removal of the inciting stimulus. Thus, smoking cessation slows the progressive loss of lung function, but the ef- fect is greatest when cessation occurs in the pre- sence of mild disease [26, 27]. Moreover, inflam- mation in the lower respiratory tract is similar among current and ex-smokers [28]. Among nor- mal smokers, cessation results in a resolution of lower respiratory tract inflammation over a num- ber of months [29, 30]. In contrast, inflammation persists, though the relative distribution of inflam- matory cells may change when a patient with COPD quits smoking [31]. This suggests that me- chanisms that lead to persistent inflammation are present in COPD. Chemotactic recruitment of in- flammatory cells by peptides derived from extra- cellular matrix, which in turn result from the ac- tion of inflammatory proteases, has been suggested as one mechanism [32–34].

Auto-immune mechanisms may also contribu- te to COPD. There is an increase in lymphocytes in the lungs of COPD patients, in particular CD8 lymphocytes [35]. In addition, an increased popu- lation of lymphocytes that are activated by elastin fragments suggests that there may be sensitization to auto-antigens in COPD [36].

Variable susceptibility

There is marked individual sensitivity to expo- sures. Thus, only about half of smokers will deve- lop COPD [37]. While the reasons for this indivi- dual susceptibility are not established, it is likely that both genetic and environmental factors con- tribute. In this context, a number of genetic fac- tors have been suggested to be related to COPD through a variety of studies [38]. Interestingly, a large number of these are related to the inflam- matory response. This suggests the concept that in- dividual heterogeneity in the inflammatory respon- se can contribute to the heterogeneity in sensitivi- ty to smoke and other exposures.

Diet has also been suggested to contribute to COPD risk, though estimating dietary exposures over the long time frames during which COPD de- velops is difficult. Nevertheless, it is possible that dietary differences also contribute to the varied susceptibility to develop COPD [39].

(4)

Other risk factors

There are several risk factors for COPD in ad- dition to exposure and conditions that are belie- ved to induce lung inflammation. Low birth we- ight, for example, is associated with low lung func- tion and increased risk of COPD in adulthood [40].

In animal models, conditions that lead to abnor- mal lung development and growth can result in emphysema [2]. This suggests that structural alte- rations of the lung can also contribute to the deve- lopment of COPD.

Tissue maintenance and COPD

The concept that inadequate tissue mainte- nance can lead to the development of COPD was suggested by Liebow [9]. Evidence for this hypo- thesis was provided by Kasahara and colleagues who demonstrated that antagonism of vascular endothelial growth factor (VEGF), a growth factor for endothelial cells, leads to endothelial cell apop- tosis and emphysema in an animal model [41].

Interestingly, this occurs in the absence of obvio- us inflammation. Kasahara and colleagues also demonstrated reduced levels of VEGF and incre- ased apoptosis in the lungs of COPD patients [42].

A number of other investigators have confirmed the presence of apoptotic cells in the lungs of COPD patients [43, 44]. In addition, several groups have shown that induction of apoptosis can cause em- physema. Yoshida and colleagues have provided a link with cigarette smoking by demonstrating that smoke, likely via oxidants, leads to activation of the intracellular stress response protein Rtp801 that, in turn, inhibits a pathway that drives VEGF production in epithelial cells. This suggests a me- chanism whereby smoke could lead to inadequate endothelial cell maintenance [45].

Repair

All tissues, including those of the lung, are capable of repair to some degree. The loss of lung tissue that defines emphysema, therefore, repre- sents a balance between tissue injury and tissue repair. In this regard, smoke can inhibit repair pro- cesses mediated by both epithelial cells and fibro- blasts [46–48].

The parenchymal cells present in the lung in COPD appear to have abnormal repair functions.

Fibroblasts cultured from the lung parenchyma of patients with COPD proliferate more slowly than controls [49, 50] and are more sensitive to inhibi- tion by smoke [51]. Cells from COPD patients are also less robust in chemotactic migration and in contraction of collagenous extracellular matrix [52], which appears to be mediated, at least in part,

by reduced sensitivity to transforming growth fac- tor-b (TGF-b), which generally induces fibroblast- mediated repair functions and by an over produc- tion of prostaglandin (PGE), which inhibits fibro- blast-mediated repair. Fibroblasts from the lungs of COPD patients also under-produce elastin [53]

and a number of mediators of repair, including hepatocyte growth factor, keratinocyte growth fac- tor [54] and fibronectin [52].

The mechanisms that lead to abnormal repair in the lung are unclear. However, it is of interest that the second largest group of genes associated with COPD are genes that are involved in repair responses (Table 1) [38]. In addition to genetic pre- disposition, an accelerated aging phenomenon has been suggested to play a role [55, 56]. In this con- text, aging of the lung is associated with an enlar- gement of airspaces that differs from COPD-asso- ciated emphysema and has been termed senile emphysema [57]. Aged animals, however, appear to be particularly susceptible to injury, and this may be a consequence of deficient repair. In this context, increased expression of markers of sene- scence has been reported in cells obtained from COPD patients, including lung fibroblasts [58, 59].

Fibrosis and COPD

While loss of repair function is a plausible mechanism that can lead to emphysema, COPD is also associated with lung fibrosis [35]. The most important lesion in the small airways associated with loss of airflow is fibrosis and narrowing. This is consistent with other fibrotic tissues where tis- sue contraction is a regular feature. The produc- tion of mediators of fibrosis, including TGF-b and fibronectin, by airway epithelial cells in response to injury has been suggested as a mechanism le- ading to airway fibrosis [60–62]. In addition, the collagen content of the pulmonary parenchyma is increased in mild emphysema [63]. This suggests the activation of repair mechanisms that may be, to some degree, overly robust.

Increased and decreased repair and cigarette smoke

Fibroblasts cultured from fibrotic tissues, in- cluding those of the lung, have been demonstra- ted to have increased “repair” functions compared to fibroblasts from controls [64, 65]. Interestingly, cigarette smoke exposure is a risk factor not only for COPD, but also for idiopathic pulmonary fibro- sis. Kanaji and colleagues (submitted) have recen- tly suggested a mechanism that can account for these disparate effects. Chronic smoke exposure can result in cellular senescence. As noted above,

(5)

cells with increased markers of senescence are pre- sent in the lungs of patients with COPD, and the associated loss of function and reduced repair co- uld contribute to the development of emphysema [55, 56]. However, in in vitro experiments, not all cells in a population undergo senescence following smoke exposure [66]. The “resistant” cells remain capable of growth. Interestingly, they grow faster and manifest more robust repair responses than controls, which resembles “fibrosis” cells. This suggests a mechanism: smoke induces senescen- ce, which has been suggested as a protective me- chanism to prevent injured cells from replicating and disrupting tissue function. Some cells, and the number may vary among individuals, acquire re- sistance to undergo senescence, and the “escaped”

cells can mediate fibrosis. A variable sensitivity to smoke-induced senescence between airway and alveolar fibroblasts could account for concurrent airway fibrosis and alveolar emphysema.

Therapeutic implications

Understanding the pathogenetic mechanisms that lead to COPD is hoped to lead to novel thera- peutic strategies. Early studies identified important mediators such as IL-8 and tumor necrosis factor a (TNF-a). Clinical trials antagonizing these media- tors, however, did not show clinical benefits altho- ugh modest effects antagonizing IL-8 are not exc- luded [67, 68]. As a result, the concept of redun- dant signaling has emerged, which has led to the- rapeutic strategies that act broadly. These include agents that are in clinical trials including: inhibi- tors of the kinase P-38, which appears to play a central signaling role for many inflammatory pa- thways; blockers of the chemokine receptor CXCR2, which mediates the response of neutro- phils to many chemotactic signals; and inhibitors

of phosphodiesterase 4, which inhibits the degra- dation of cAMP levels in many inflammatory cells and thereby reduces inflammation. One member of the last family, roflumilast [69, 70] has recently been approved as a therapy for COPD.

Repair of the lung is also a reasonable goal.

Retinoic acid, which stimulates alveolar wall growth in neonatal animals [71], was shown to sti- mulate repair of alveolar wall in emphysema in animal models. Two clinical trials in COPD pa- tients with all-trans retinoic acid were without benefit [72, 73]. However, trials with an agonist selective for the RAR-g receptor, which appears to mediate the repair response, have been initiated.

Similarly, trials with stem cell infusions have been undertaken.

References

1. Global Strategy for Diagnosis. Management and Prevention of COPD. 2009.

2. Shapiro S.D., Reilly J.J.J., Rennard S.I. Chronic Bronchitis and Emphysema. In: Mason R.J., Martin T.R., King T.E., Jr., Schraufna- gel D.E., Murray J.F., Jay A. (eds.). Nadel Textbood of Respiratory Medicine, Vol. I. Saunders, Philadelphia 2010: 919–967.

3. Han M.K. et al. COPD Phenotypes: The Future of COPD. Am. J.

Respir. Crit. Care. Med. 2010; 182: 598–604.

4. Rennard S.I., Vestbo J. The many „small COPDs”: COPD should be an orphan disease. Chest 2008; 134: 623–627.

5. Niewoehner D. Structure-Function Relationships: the Patho- physiology of Airflow Obstruction. In: Stockley R., Rennard S., Rabe K., Celli, B. (eds.). Chronic Obstructive Pulmonary Dis- ease. Blackwell, Malden 2007: 3–19.

6. Orie N.G., Sluiter H.J., DeVries K., Tammeling G.J., Witkop, J.

The host factor in bronchitis. In: Orie, N.G.M., Sluiter, H.J.

(eds.). Bronchitis, An International Symposium. Assen, Nether- lands 1961: 43–59.

7. Fletcher C., Peto R. The natural history of chronic airflow ob- struction. Br. Med. J. 1977; 1: 1645–1648.

8. Laurell C.B., ErikssonS. The electrophoretic alpha 1-globulin pattern of serum in alpha 1-antitrypsin deficiency. Scand J.

Clin. Lab. Invest. 1963; 15: 132–140.

9. Liebow A.A. Pulmonary emphysema with special reference to vascular changes. Am. Rev. Respir. Dis. 1959; 80: 67–93.

10. Salvi S.S., Barnes P.J. Chronic obstructive pulmonary disease in non-smokers. Lancet 2009; 374: 733–743.

11. Barnes P.J. The cytokine network in asthma and chronic ob- structive pulmonary disease. J. Clin. Invest. 2008; 118: 3546–

–3556.

12. Mio T., Romberger D.J., Thompson A.B. et al. Cigarette smoke induces interleukin-8 release from human bronchial epithelial cells. Am. J. Respir. Crit. Care Med. 1997; 155: 1770–1776.

13. Kew R.R., Ghebrehiwet B., Janoff A. Cigarette smoke can acti- vate the alternate pathway of complement in vitro by modify- ing the third component of complement. J. Clin. Invest. 1985;

75: 1000–1007.

14. Robbins R.A., Nelson K.J., Gossman G.L., Koyama S., Rennard S.I. Complement activation by cigarette smoke. Am. J. Physiol.

1991; 260: L254–L259.

15. Zhang J., Kew R.R. Identification of a region in the vitamin D- binding protein that mediates its C5a chemotactic cofactor func- tion. J. Biol. Chem. 2004; 279: 53282–53287.

16. Robbins R.A., Gossman G.L., Nelson K.J. et al. Inactivation of chemotactic factor inactivator by cigarette smoke. A potential mechanism of modulating neutrophil recruitment to the lung.

Am. Rev. Respir. Dis. 1990; 142: 763–768.

17. Park J.A., He F., Martin L.D., Li Y., Chorley B.N., Adler K.B.

Human neutrophil elastase induces hypersecretion of mucin from well-differentiated human bronchial epithelial cells in vit- ro via a protein kinase C{delta}-mediated mechanism. Am. J.

Pathol. 2005; 167: 651–661.

Table 1.

Genetic and COPD

ACE ADRBZ EPHX1 GSTP1 IL13 IL1B IL4 IL6 LTA MMP9

SERPINA3 SOD3 TGFB1 TIMP2 TVF GSTM1 GSTT ADAM33 TLR4

(6)

18. Shao M.X., Nadel J.A. Neutrophil elastase induces MUC5AC mucin production in human airway epithelial cells via a cas- cade involving protein kinase C, reactive oxygen species, and TNF-alpha-converting enzyme. J. Immunol. 2005; 175: 4009–

–4016.

19. Voynow J.A., Fischer B.M., Malarkey D.E. et al. Neutrophil elastase induces mucus cell metaplasia in mouse lung. Am. J.

Physiol. Lung Cell Mol. Physiol. 2004; 287: L1293–L1302.

20. MacNee W. Pulmonary and systemic oxidant/antioxidant im- balance in chronic obstructive pulmonary disease. Proc. Am.

Thorac. Soc. 2005; 2: 50–60.

21. Decramer M., Rutten-van Mölken M., Dekhuijzen P.N. et al.

Effects of N-acetylcysteine on outcomes in chronic obstructive pulmonary disease (Bronchitis Randomized on NAC Cost-Utili- ty Study, BRONCUS): a randomised placebo-controlled trial.

Lancet 2005; 365: 1552–1560.

22. Massaro D., Massaro G.D., Baras A., Hoffman E.P., Clerch L.B.

Calorie-related rapid onset of alveolar loss, regeneration, and changes in mouse lung gene expression. Am. J. Physiol. Lung Cell Mol. Physiol. 2004; 286: L896–L906.

23. Petrache I., Natarajan V., Zhen L. et al. Ceramide upregulation causes pulmonary cell apoptosis and emphysema-like disease in mice. Nat. Med. 2005; 11: 491–498.

24. Carp H., Janoff A. In vitro suppression of serum elastase inhibi- tory capacity by reactive oxygen species generated by phagocy- tosing polymorphonuclear leukocytes. J. Clin. Invest. 1979; 63:

793–797.

25. Lange P., Parner J., Vestbo J., Schnor P., Jensen G. A 15-year follow-up study of ventilatory function in adults with asthma.

New Engl. J. Med. 1998; 339: 1194–1200.

26. Anthonisen N.R., Connett J.E., Kiley J.P. et al. Effects of smok- ing intervention and the use of an inhaled anticholinergic bron- chodilator on the rate of decline of FEV1. JAMA 1994; 272:

1497–1505.

27. Kohansal R., Martinez-Camblor P., Agustí A., Buist A.S., Man- nino D.M., Soriano J.B. et al. The natural history of chronic airflow obstruction revisited: an analysis of the Framingham offspring cohort. Am. J. Respir. Crit. Care Med. 2009; 180: 3–10.

28. Rutgers S.R., Postma D.S., ten Hacken N.H. et al. Ongoing air- way inflammation in patients with COPD who do not currently smoke. Thorax 2000; 55: 12–18.

29. Skold C.M., Hed J., Eklund A. Smoking cessation rapidly re- duces cell recovery in bronchoalveolar lavage, while alveolar macrophage fluorescence remains high. Chest 1992; 101: 989–

–995.

30. Swan G.E., Hodgkin J.E., Roby T. et al. Reversibility of airways injury over a 12-month period following smoking cessation.

Chest 1992; 101: 607–612.

31. Willemse B.W., ten Hacken N.H., Rutgers B. et al. Effect of 1-year smoking cessation on airway inflammation in COPD and asymptomatic smokers. Eur. Respir. J. 2005; 26: 835–845.

32. Hunninghake G.W., Davidson J.M., Rennard S. et al. Elastin fragments attract macrophage precursors to diseased sites in pulmonary emphysema. Science 1981; 212: 925–927.

33. Senior R.M., Griffin G.L., Meacham R.P. Chemotactic activity of elastin-derived peptides. J. Clin. Invest. 1980; 66: 859–862.

34. Gaggar A., Jackson P.L., Noerager B.D. et al. A novel proteolytic cascade generates an extracellular matrix-derived chemoattrac- tant in chronic neutrophilic inflammation. J. Immunol. 2008;

180: 5662–5669.

35. Hogg J.C., Chu F., Utokaparch S. et al. The nature of small- airway obstruction in chronic obstructive pulmonary disease.

N. Engl. J. Med. 2004; 350: 2645–2653.

36. Lee S.H., Goswami S., Grudo A. et al. Antielastin autoimmunity in tobacco smoking-induced emphysema. Nat. Med. 2007; 13:

567–569.

37. Rennard S.I., Vestbo J. COPD: The dangerous underestimate of 15%. Lancet 2006; 367: 1216–1219.

38. Castaldi P.J., Cho M.H., Cohn M. et al. The COPD genetic asso- ciation compendium: a comprehensive online database of COPD genetic associations. Hum. Mol. Genet. 2009; 19: 526–

–534.

39. McKeever T.M., Lewis S.A., Cassano P.A. et al. Patterns of di- etary intake and relation to respiratory disease, forced expirato- ry volume in 1 s, and decline in 5-y forced expiratory volume.

Am. J. Clin. Nutr. 2010; 92: 408–415.

40. Barker D.J., Godfrey K.M., Fall C. et al. Relation of birthweight and childhood respiratory infection to adult lung function and death from chronic obstructive airways disease. BMJ 1991; 303:

671–675.

41. Kasahara Y., Tuder R.M., Taraseviciene-Stewart L. et al. Inhibi- tion of VEGF receptors causes lung cell apoptosis and emphy- sema. J. Clin. Invest. 2000; 106: 1311–1319.

42. Kasahara Y., Tuder R.M., Cool C.D. et al.Endothelial cell death and decreased expression of vascular endothelial growth factor and vascular endothelial growth factor receptor 2 in emphyse- ma. Am. J. Respir. Crit. Care Med. 2001; 163: 737–744.

43. Taraseviciene-Stewart L., Voelkel N.F. Molecular pathogenesis of emphysema. J. Clin. Invest. 2008; 118: 394–402.

44. Plataki M., Tzortzaki E., Rytila P. et al. Apoptotic mechanisms in the pathogenesis of COPD. Int. J. Chron. Obstruct. Pulmon.

Dis. 2006; 1: 161–171.

45. Yoshida T., Mett I., Bhunia A.K. et al. RTP801, a suppressor of mTOR signaling, is an essential mediator of cigarette smoke- induced pulmonary injury and emphysema. Nature Medicine 2010; 16: 767–773.

46. Cantral D.E., Sisson J.H., Veys T., Rennard S.I., Spurzem, J.R.

Effects of cigarette smoke extract on bovine bronchial epithelial cell attachment and migration. Am. J. Physiol. 1995; 268: L723–

–L728.

47. Carnevali S., Nakamura Y., Mio T. et al. Cigarette smoke extract inhibits fibroblast-mediated collagen gel contraction. Am. J.

Physiol. 1998; 274: L591–L598.

48. Nakamura Y., Romberger D.J., Tate L. et al. Cigarette smoke inhibits lung fibroblast proliferation and chemotaxis. Am. J.

Respir. Crit. Care Med. 1995; 151, 1497–1503.

49. Holz O., Zühlke I., Jaksztat E. et al. Lung fibroblasts from pa- tients with emphysema show a reduced proliferation rate in culture. Eur. Respir. J. 2004; 24: 575–579.

50. Noordhoek J.A., Postma D.S., Chong L.L. et al. Different prolif- erative capacity of lung fibroblasts obtained from control sub- jects and patients with emphysema. Exp. Lung Res. 2003; 29:

291–302.

51. Nobukuni S., Watanabe K., Inoue J. et al. Cigarette smoke in- hibits the growth of lung fibroblasts from patients with pulmo- nary emphysema. Respirology 2002; 7: 217–223.

52. Togo S., Holz O., Liu X. et al. Lung fibroblast repair functions in patients with chronic obstructive pulmonary disease are al- tered by multiple mechanisms. Am. J. Respir. Crit. Care Med.

2008; 178: 248–260.

53. Plantier L., Rochette-Egly C., Goven D. et al. Dysregulation of elastin expression by fibroblasts in pulmonary emphysema: role of cellular retinoic acid binding protein 2. Thorax 2008; 63:

1012–1017.

54. Plantier L., Marchand-Adam S., Marchal-Sommé J. et al. Defect of hepatocyte growth factor production by fibroblasts in human pulmonary emphysema. Am. J. Physiol. Lung Cell Mol. Physi- ol. 2005; 288: L641–L647.

55. Aoshiba K., Nagai A. Senescence hypothesis for the pathoge- netic mechanism of chronic obstructive pulmonary disease.

Proc. Am. Thorac. Soc. 2009; 6: 596–601.

56. MacNee W., Tuder R.M. New paradigms in the pathogenesis of chronic obstructive pulmonary disease I. Proc. Am. Thorac.

Soc. 2009; 6: 527–531.

57. Fukuchi Y. The aging lung and chronic obstructive pulmonary disease: similarity and difference. Proc. Am. Thorac. Soc. 2009;

6: 570–572.

58. Müller K.C., Welker L., Paasch K. et al. Lung fibroblasts from patients with emphysema show markers of senescence in vitro.

Respir. Res. 2006; 7: 32.

59. Tsuji, T., Aoshiba, K., Nagai, A. Alveolar cell senescence in patients with pulmonary emphysema. Am. J. Respir. Crit. Care Med. 2006; 174: 886–893.

60. Shoji S., Rickard K.A., Ertl R.F. et al. Bronchial epithelial cells produce lung fibroblast chemotactic factor: Fibronectin. Am. J.

Respir. Cell Molec. Biol. 1989; 1: 13–20.

61. Takizawa H., Tanaka M., Takami K. et al. Increased expression of transforming growth factor-beta1 in small airway epithelium from tobacco smokers and patients with chronic obstructive pulmonary disease (COPD). Am. J. Respir. Crit. Care Med. 2001;

163: 1476–1483.

62. Holgate, S. Epithelial damage and response. Clin. Exp. Allergy 2000; 30 (Suppl.): 137–141.

63. Martin-Mosquero C., Peces-Barba G., Rubio M.L. et al. Increased collagen deposition correlated with lung destruction in human emphysema. Histol. Histopathol. 2006; 21: 823–828.

64. Noble, P.W. Idiopathic pulmonary fibrosis: natural history and prognosis. Clin Chest Med 2006; 27 (Suppl. 1): S11–S16.

65. King T.E. Jr., Schwarz M.I., Brown K. et al. Idiopathic Pulmo- nary Fibrosis. Relationship between histopathologic features

(7)

and mortality. Am. J. Respir. Crit. Care Med. 2001; 164: 1025–

–1032.

66. Nyunoya T., Monick M.M., Klingelhutz A. et al. Cigarette smoke induces cellular senescence. Am. J. Respir. Cell Mol. Biol. 2006;

35: 681–8.

67. Rennard S.I., Fogarty C., Kelsen S. et al. The Safety and Effica- cy of Infliximab in Moderate-To-Severe Chronic Obstructive Pulmonary Disease. Am. J. Respir. Crit. Care Med. 2007; 175:

926–934.

68. Mahler D.A., Huang S., Tabrizi M., Bell G.M. Efficacy and safe- ty of a monoclonal antibody recognizing interleukin-8 in COPD:

a pilot study. Chest 2004; 126: 926–934.

69. Fabbri L.M., Calverley P.M., Izquierdo-Alonso J.L. et al. Roflu- milast in moderate-to-severe chronic obstructive pulmonary

disease treated with longacting bronchodilators: two ran- domised clinical trials. Lancet 2009; 374: 695–703.

70. Calverley P.M., Rabe K.F., Goehring U.M. et al. Roflumilast in symptomatic chronic obstructive pulmonary disease: two ran- domised clinical trials. Lancet 2009; 374: 685–694.

71. Massaro G., Massaro D. Retinoic acid treatment abrogates elastase-induced pulmonary emphysema in rats. Nature Medi- cine 1997; 3: 675–677.

72. Mao J.T., Goldin J.G., Dermand J. et al. A pilot study of all- trans-retinoic acid for the treatment of human emphysema. Am.

J. Respir. Crit. Care Med. 2002; 165: 718–723.

73. Roth M.D., Connett J.E., D’Armiento J.M. et al. Feasibility of retinoids for the treatment of emphysema study. Chest 2006;

130: 1334–1345.

Cytaty

Powiązane dokumenty

Dyskusyjne jest jednak stwierdzenie autorek, że szczepienia przeciw grypie są zalecane u chorych na ciężką postać POChP, zwłaszcza w podeszłym wieku [1].. Jest to sprzeczne

Non-invasive positive pressure ventilation for treatment of respiratory failure due to exacerbations of chronic obstructive pulmonary disease.. Cochrane Database of Systematic

This paper explores the natural history of chronic obstructive pulmonary disease (COPD), including our present understand- ing of COPD risk factors, phenotypes, and burden, along

Chronic obstructive pulmonary disease (COPD) and obstructive sleep apnoea (OSA) are the two most common chronic respiratory disorders in the adult population.. The prevalence of COPD

The aim of our study was to determine the relationship between the severity of COPD and the occurrence of chronic alveolar hypoventilation in obese patients with moderate to severe

Koszt chorego na POChP jest natomiast 5-krotnie niższy niż chorego, u którego stwierdza się astmę i POChP.. Wykazano wyraźną korelację pomiędzy kosz- tem a stopniem

In The Copenhagen City Heart Study, where we followed a large cohort of almost 15 000 individuals selected from the general population including patients with COPD, we could show

Wykazano także, że komórki nabłon- ka oddechowego wykazują podwyższoną ekspre- sję białka zapalnego IP-10 (inflammatory protein-10), które jest silnym czynnikiem chemotaktycznym