Download as pdf or txt
Download as pdf or txt
You are on page 1of 5

232 Endocrine, Metabolic & Immune Disorders - Drug Targets, 2007, 7, 232-236

Tetracyclines and Pulmonary Inflammation


*
S. Rempe, J.M. Hayden, R.A. Robbins and J.C. Hoyt

Research Service, Carl T. Hayden VA Medical Center Phoenix, AZ, University of Arizona, Tucson, AZ, USA

Abstract: Tetracycline and its derivatives, such as chlortetracycline, oxytetracycline, minocycline, doxycycline, methacy-
cline and lymecycline, are naturally occurring or semi-synthetic polyketide compounds that exhibit a well known broad-
spectrum antibacterial activity that interferes with prokaryotic protein synthesis at the ribosome level. In addition to this
well known antibacterial activity these compounds also exhibit a variety of additional, less well known properties. Among
them are separate and distinct anti-inflammatory properties. Tetracycline and related compounds have been shown to be
effective chemotherapeutic agents in a wide variety of chronic inflammatory diseases and conditions. These include pe-
riodontitis, rosacea, acne, auto-immune diseases such as rheumatoid arthritis and protection of the central nervous system
against trauma and neurodegenerative diseases such as stroke, multiple sclerosis and Parkinson disease. Tetracycline and
related compounds appear to be beneficial for treatment of several chronic inflammatory airway diseases. Among them
are asthma, bronchiectasis, acute respiratory distress syndrome, chemical induced lung damage and cystic fibrosis. The
clinical use of tetracycline-type drugs in treatment of chronic airway inflammation is becoming a topic of intense interest.
Recent findings in this area have led to an understanding of the myriad physiological, cellular and molecular mechanisms
of the inflammatory response and how this response may be controlled to limit damage to host cells and tissues. This re-
view presents a brief summary of the recent research in the area of tetracycline and its derivatives in control of pulmonary
inflammation.
Key Words: Tetracycline, doxycycline, lung, inflammation.

INTRODUCTION well known antibiotic or antibacterial actions. Presently a


subantimicrobial dose of doxycycline is an approved treat-
Tetracyclines, discovered in 1948, are a group of poly-
ment for inflammation associated with periodontal disease
ketide antibiotic compounds, produced in the soil by several
[5].
members of the bacterial genus Streptomyces. In 1954 the
first member of the tetracycline family to be characterized, Since this first demonstration of anti-inflammatory action
chlortetracycline Fig. (1), was isolated from fermentation in periodontal disease, the tetracycline and related com-
broths of Streptomyces aureofacians [1]. Clinical studies pounds have been examined in additional clinical settings.
with this newly discovered antibiotic demonstrated advan- Inflammation and underlying tissue destruction caused by
tages over penicillin such as a greater spectrum of antibacte- two potentially disfiguring diseases acne vulgaris and ro-
rial activity and fewer adverse effects. Since the discovery of sacea has been shown to be dramatically reduced by low
chlortetracycline (1) other naturally occurring derivatives dose doxycycline therapy [6]. O’Dell et al. [7] demonstrated
such as tetracycline (2) and oxytetracycline (3) have also that doxycycline with methotrexate was a superior treatment
been isolated. Modification of these naturally occurring anti- over methotrexate alone in the treatment of early seroposi-
biotics has lead to the development of a series of clinically tive rheumatoid arthritis patients. However, little benefit was
useful semi-synthetic tetracycline derivatives Fig. (1). Among observed if treatment was delayed for a substantial time. It is
these derivatives are minocycline (4), doxycycline (5), thought that anti-inflammatory action of doxycycline is
methacycline (6) and lymecycline (7). These semi-synthetic caused by inhibition of the collagenase activity of one or
tetracycline derivatives often exhibit increased absorption, more matrix metalloproteinase enzymes. In a similar study
decreased bacterial resistance and increased efficacy over the lymecycline was demonstrated to be of possible benefit in
naturally occurring analogs [2] The biosynthesis, chemical the treatment of reactive arthritis [8]. Minocycline has re-
and physical properties of the tetracycline compounds has ceived attention as a possible protective agent in neurologi-
been well described in several review articles [2,3] and will cal injury and disease in part because of its ability to diffuse
not be covered in this brief review. into the central nervous system and reach therapeutic con-
centrations. Minocycline has been shown to decrease proin-
During the 1980s Golub et al. [4] observed that mino-
flammatory cytokine production and decrease neural cell
cycline decreased gingival collagenase levels and retarded death leading to improved functional outcomes of spinal
alveolar bone loss in germ-free diabetic rats giving the first
cord injured rats [9]. Other studies have shown that mino-
indication that tetracycline-type compounds exhibit a second
cycline reduces inflammation in the brain by inhibition of
mode of action, anti-inflammatory action, in addition to their
matrix metalloproteinases and microglial activation [10].
Other neurological diseases that may benefit from the anti-
inflammatory action of minocycline include stroke, multiple
*Address correspondence to this author at the Carl T. Hayden VA Medical
Center, Research (RS/151), Building 27, 650 East Indian School Road, sclerosis, Parkinson disease, amyotrophic lateral sclerosis,
Phoenix, AZ 85012, USA; Tel: (602) 277-5551; Fax: (602) 212-2047; Alzheimer disease and Huntington disease [10]. Using a sim-
E-mail: jeff.hoyt2@va.gov ian immunodeficiency model of human immunodeficiency

1871-5303/07 $50.00+.00 © 2007 Bentham Science Publishers Ltd.


Tetracyclines and Pulmonary Inflammation Endocrine, Metabolic & Immune Disorders - Drug Targets, 2007, Vol. 7, No. 3 233

R4 R2 R3 R1 R
OH
6 5 4
8 7 3
D C B A O
9 10 11 12 2
1

OH HN
OH O OH O R5

R R1 R2 R3 R4 R5
1 Chlortetracycline -N(CH3)2 -H -CH3 -H -Cl -H
2 Tetracycline -N(CH3)2 -H -CH3 -H -H -H
3 Oxytetracycline -N(CH3)2 -OH -CH3 -OH -H -H
4 Minocycline -N(CH3)2 -H -H -H -N(CH3)2 -H
5 Doxycycline -N(CH3)2 -OH --CH3 -H -H -H
6 Methacycline -N(CH3)2 -OH =CH3 -- -H -H
7 Lymecycline -N(CH3)2 -H -CH3 -OH -H -N--Methylene Lysine
8 CMT-8 -H -H -H -H -H -H

Fig. (1).

virus (HIV) central nervous system disease Zink et al. [11] ing tetracycline and its derivatives, used in the treatment of
have demonstrated that minocycline treatment reduced se- pulmonary inflammation.
verity of encephalitis, suppressed viral load and decreased
Effect of Tetracyclines on Pulmonary Inflammation
expression of various inflammatory markers. This fascinat-
ing work indicates that minocycline may be a possible anti- The inflammatory response consists of a diverse, com-
inflammatory therapy for HIV disease. plex series of chemical messengers and cellular events that
can occur throughout the lung. The anti-inflammatory effects
PULMONARY INFLAMMATION AND TETRACY-
CLINES of tetracyclines may occur at a variety of different sites and
in differing ways and tetracycline-type drugs exhibit good
Common airway diseases are the result of injury to the tissue penetration exceeding therapeutic concentrations [13-
lung, are characterized by changes in lung function, tissues 15] making these drugs possible anti-inflammatory agents.
and fluids and are associated with chronic inflammation.
Inflammation is a continuous process in lung tissues. It
Current thinking suggests that this inflammation is caused by
serves as a host defense to protect the lungs from infectious
chemical attractants, such as cytokines and lipid factors, that
lead to the migration and accumulation of inflammatory cells and noxious agents. However, it is a thin line between in-
flammation protecting the lungs and inflammation actually
in the lung tissues and airspaces. The type of injury (e.g.
causing tissue destruction [16]. Matrix metalloproteinases,
infectious, allergy or smoke/chemical inhalation) will deter-
nitric oxide and neutrophil elastases are some of the many
mine the pattern or type of cell infiltration [12]. This cell
potentially damaging mediators found in inflamed lung tis-
migration is followed by lung tissue inflammation and dam-
sues.
age. Despite considerable research, the role of these chemi-
cal attractants and conditions that control their function in Matrix Metalloproteinases
airway diseases remain unclear. This brief review covers
research findings concerning tetracycline-type antibiotics Matrix metalloproteinases (MMP) are a family of zinc
and their effect on inflammatory pulmonary diseases. dependent endopeptidase isozymes which are able to degrade
the extracellular matrix and are responsible for tissue remod-
Distribution of Drugs in Lung Tissues eling and damage in many situations. They are involved in
wound healing, cancer progression and have also been im-
Whether antibiotic compounds are used to treat bacterial
infections or to treat inflammation their ultimate disposition plicated in causing lung damage in various lung diseases.
There are more than 20 MMPs known. In the lung two MMP
in tissues and cells must be considered. Oral or systemic
isozymes, MMP-2 (gelatinase A) and MMP-9 (gelatinase B),
administration of antibiotics leads to their accumulation
have roles in lung tissue remodeling and damage. Both
through out the body including lung tissues. However, many
isozymes are released from non-inflammatory cells, while
factors or mechanisms effect the ultimate disposition of these
inflammatory cells mainly produce MMP-9 [17-19]. These
agents in the lungs. Among these mechanisms are transport
of the agent into the respiratory tract tissues from the cardio- two isozymes degrade gelatin and collagen localized to the
endothelial basement membrane and basement membranes
vascular system, host-related factors such as inflammation
of smooth muscle cells of the intima and media. MMPs have
and mechanical injury and antibiotic-related factors such as
been implicated in the pathogenesis of many lung disorders.
molecular weight, hydrophobicity, degree of protein binding,
Increased levels of MMPs have been found in bronchoalveo-
biotransformation or destruction of the drug, the speed of
lar lavage (BAL) fluid of acute respiratory distress syndrome
absorption and excretion and the final concentration achieved
[13]. These factors influence the choice of antibiotic, includ- (ARDS) patients and it has been suggested that they play a
234 Endocrine, Metabolic & Immune Disorders - Drug Targets, 2007, Vol. 7, No. 3 Rempe et al.

major role in ARDS and in its development of capillary leak Guignabert et al. [32] used guinea pigs to induce respira-
syndrome [20,21]. MMPs also play a role in chronic in- tory epithelial lesions via intra-tracheal administration of
flammation in cystic fibrosis (CF). Sagel et al. [22] showed sulfur mustard. Sulfur mustard, a vesicant, is a chemical war-
that induced sputum levels of MMP-9 were significantly fare agent that causes severe blistering when it makes contact
increased in children with CF compared with healthy con- with tissues and cells. They demonstrated increased levels of
trols. They also showed that the ratio of MMP-9 to its inhibi- gelatinase in BAL fluid 24 hours after exposure by zy-
tor, TIMP-1, was higher in cystic fibrosis patients compared mographic analysis of biotinylated substrate degradation.
with controls and that there was a significant inverse rela- Treatment with subcutaneous administration of doxycycline
tionship between MMP-9 and FEV1. Similarly Ratjen et al. (30 mg/kg) 3 hours before exposure to sulfur mustard de-
[23] showed increased levels of MMP-8 and -9 in pulmonary creased gelatinase activity, inflammation and epithelial le-
lavage fluid of children with cystic fibrosis. Excessive levels sions. Rappenau et al. [33] used the human bronchial epithe-
of MMP-8 and -9 have also been found in patients with hos- lial cell line 16HBE14o- to demonstrate that a combination
pital-acquired pneumonia [24] and an abundance of MMPs of N-acetyl cysteine and doxycycline provided improved
have been found in sarcoid granulomas [25]. protection against sulfur and nitrogen mustard vesicants over
each separate treatment.
Given the role of MMPs in these and many other lung
disorders it appears crucial to apply some therapeutic inter- Nitric Oxide
vention. Golub et al. [4] demonstrated that minocycline in-
hibited collagenase (one form of MMP). Since then tetracy- Nitric oxide (NO) is constitutively produced at low basal
levels by many pulmonary cells by the constitutive nitric
cline and several of its derivatives have been shown to in-
oxide synthase (NOS) type I or type II isozymes or can be
hibit the activity of a variety of MMP isozymes indicating a
produced in larger amounts during inflammation by the in-
possible treatment for inflammation and inflammatory lung
ducible form of NOS (iNOS or type II NOS). NOS catalyzes
diseases. Recent work by Garcia et al. [26] examined the
oxidation of L-arginine to NO and L-citrulline. NO is a free
mechanism of doxycycline inhibition of matrilysin (MMP-7)
using deuterium exchange mass spectroscopy. Matrilysin radical which under physiological conditions is responsible
for controlling the smooth muscle tone in vessels and air-
inhibition is apparently not due to binding of doxycycline to
ways. It also acts as a proinflammatory mediator. NO can
the enzyme’s active site but rather by its interaction with
react with superoxide produced by activated macrophages to
enzyme bound zinc or calcium atoms that are required for
produce peroxynitrite, a powerful oxidizing agent, which
structural stability of the enzyme. Binding of doxycycline to
may cause wide spread, severe lung damage. NO production
these metal atoms apparently destabilizes the enzyme struc-
ture leading to loss of activity. has been implicated in the pathogenesis of many inflamma-
tory lung diseases including ARDS, reactive airway disease
Lee et al. [27] examined the effect of doxycycline on and cystic fibrosis. It is also believed to be involved in silica-
airway hyper-responsiveness and inflammatory mediators in induced lung disease [34]. Genovese et al. [35] examined the
toluene diisocyanate-induced (TDI) asthma using a murine role of NO in bleomycin-induced lung disease. Mice that
system. TDI is a common cause of occupational asthma lacked the iNOS gene and that had received bleomycin de-
which is characterized by airway hyper-responsiveness and veloped less severe lung injury, namely decreased edema and
inflammation. Doxycycline (20 mg/kg) was administered by PMN induced inflammation, leading to a decrease in rate of
gavage daily for six days before TDI challenge. Doxycycline mortality.
was found to reduce MMP-9 mRNA and protein levels, de-
The effect of doxycycline on NO production by LA4
crease the number of lymphocytes, eosinophils and neutro-
murine lung epithelial cells has been examined [36]. Doxy-
phils in airway lumen and reduce airway hyper-responsive-
cycline decreased iNOS catalyzed production of NO via a
ness compared to untreated controls.
reduction in iNOS protein and iNOS mRNA. In addition
Palei et al. [28] examined the role of MMP-2 and MMP- doxycycline decreased production of p38 MAP kinase which
9 during acute pulmonary embolism (APE) in rats. Doxycy- in turn decreased the stability of iNOS mRNA ultimately
cline, 30 mg/kg injected i.v., was found to decrease MMP-9 leading to decreased production of NO. In similar work
activity but not MMP-2 activity after embolization. MMP-9 D’Agustino et al. [37] found that doxycycline treatment re-
is involved in regulation of vascular tone through endothelin- duced mortality arising from endotoxemia in a murine
1 release [29]. It is possible that APE enhanced production model. Doxycycline was found to reduce synthesis of iNOS
of MMP-9 is responsible for increased activation of endo- in endotoxin-stimulated macrophages by an IL-10 independ-
thelin-1 and that doxycycline inhibition of MMP-9 may ac- ent mechanism, and the reduction of NO production after
count for endothelin-1 mediated hemodynamic changes dur- exposure to endotoxin may have reduced inflammatory dam-
ing APE [30]. age from septic shock.
The severity of bronchiectasis has been found to correlate Neutrophil Elastase
with the levels of neutrophil collagenase (MMP-8) contained
in bronchoalveolar lavage (BAL) fluid [31]. A correlation Neutrophil elastase (NE) is a serine protease located in
azurophil granules of leukocytes. NE degrades elastin, an
between the levels of active MMP-8 and severity of lung
extracellular matrix protein of the lung, which gives lungs
tissue damage in bronchiectasis was confirmed. Exposure to
their elastic recoil. This degradation, if uncontrolled, may
doxycycline inhibited MMP-8 activity further suggesting
cause severe inflammation and tissue damage. NE has been
that it may be a possible therapeutic intervention to prevent
implicated in various inflammatory lung diseases including
long term tissue damage in bronchiectasis.
Tetracyclines and Pulmonary Inflammation Endocrine, Metabolic & Immune Disorders - Drug Targets, 2007, Vol. 7, No. 3 235

ARDS [38], chronic obstructive pulmonary disease (COPD)/ rum and BAL fluid levels of the aforementioned cytokines
emphysema [39] and cystic fibrosis [40]. An imbalance be- and NE.
tween NE and its inhibitors, such as alpha-1-antitrypsin,
seems to play a key role in these diseases [41]. Tetracycline Carney et al. [47] used endotoxin-induced ARDS to ex-
derivatives have been reported to prevent MMP-catalyzed amine the effect of COL-3 on lung function and activity of
degradation of NE inhibitors protecting the extracellular ma- MMP-2 and MMP-9. Pretreatment with COL-3 decreased
trix from extensive destruction [42]. sequestration of neutrophils and monocytes in lung tissues,
reduced the wet/dry weight ratio of the lung and decreased
Effect on Cytokines the destructive proteolytic activity of both MMP-2 and
MMP-9.
Monocyte chemoattractant protein-1 (MCP-1) is a major
pro-inflammatory chemokine found in chronic pulmonary Although CMTs do not exhibit antibacterial activity,
disease and is a potent chemoattractant for monocytes. Using there is evidence that they may be effective antifungal agents
A549 human lung epithelial cells Raza et al. [43] were able [48,49]. The chemically modified tetracycline used these
to show that doxycycline, 30 g/ml, decreased production of studies, CMT-3, was found to be a more effective fungicide
MCP-1. Expression of MCP-1 mRNA was found to be de- against filamentous fungi than against yeasts. In contrast, the
pressed by doxycycline treatment. In a similar fashion inter- often used fungistatic compound amphotericin B was found
leukin-8 (IL-8) production was also determined to be de- to be effective against most filamentous fungi and yeast
pressed by doxycycline treatment of stimulated A549 epithe- tested. The viability of two filamentous fungi that can cause
lial cells (Hoyt, unpublished results). Tigecycline is the first lung infections, Aspergillus fumigatus and Rhizopus sp. was
clinically available drug in a new glycylcycline class of anti- reduced approximately 60% after treatment with CMT-3 on
biotics. Tigecycline is a derivative of minocycline with a SABHI agar; however, a greater viability of both organisms
- - - -
(CH3)3C NH-CH2 C(O) NH moiety on position 9 of the D was seen in similar experiments using amphotericin B.
ring. Salvatore et al.1 examined the effect of tigecycline in a
murine model of Mycoplasma pneumoniae induced pneumo- CONCLUSIONS
nia. In BAL fluid from infected mice tigecycline was found
The use of tetracycline and its derivatives in the treat-
to decrease concentrations of proinflammatory cytokines and ment of a variety of inflammatory conditions beyond the
chemokines including TNF-, IFN-, IL-1, GM-CSF,
lung is becoming increasingly apparent. The therapeutic po-
MCP-1 and others relative to uninfected controls. tential of this class of antibiotic in pulmonary inflammation
Chemically Modified Tetracyclines is especially important in chronic conditions such as bron-
chiectasis, COPD, cystic fibrosis, and many others. The ac-
Tretment of chronic inflammation often requires long- tion of these compounds is multi-factorial in nature and ef-
term exposure to various pharmacologic agents. Various fects numerous, diverse parts of the inflammatory cascade
tetracycline derivatives have been demonstrated to possess such as MMPs, neutrophil elastase, NO and pro-inflammatory
anti-inflammatory properties in addition to their better chemokines for example. In many cases the exact mecha-
known antibacterial activities. Thus, long term exposure to nism by which tetracycline-type compounds exert their anti-
tetracycline derivatives may lead to accumulation of tetracy- inflammatory effects remains undiscovered and demon-
cline-resistant bacteria ultimately leading to cessation of strates great promise for future therapy.
treatment. Realization that antibiotic activity of tetracycline
resides only in one portion of the molecule lead Golub et al. ABBREVIATIONS
[44] to chemically modify the basic tetracycline structure in
APE = Acute Pulmonary Embolism
order to abolish this activity. Removal of the dimethylamino
group from position 4 of the A-ring Fig. (1) abolished anti- ARDS = Acute Respiratory Distress Syndrome
bacterial activity [44] and lead to the synthesis of a series of
BAL = Bronchoalveolar Lavage
chemically modified tetracyclines or CMTs [45]. The vari-
ous CMTs have been examined in a variety of systems for CF = Cystic Fibrosis
anti-inflammatory activity.
CMT = Chemically Modified Tetracycline
Steinberg, et al. [46] examined the effect of COL-3, also
COPD = Chronic Obstructive Pulmonary Disease
known as CMT-3 (8), on septic shock and ARDS in a por-
cine model. Occlusion of the superior mesenteric artery was HIV = Human Immunodeficiency Virus
used to induce sepsis. As septic shock developed, lung tissue
IL = Interleukin
revealed damage typical of ARDS, accompanied by an ob-
served increase in concentration of plasma IL-1 and IL-6 and iNOS = Inducible Nitric Oxide Synthase Isozyme
BAL IL-6, IL-8, IL-10 and NE. Pretreatment with COL-3
prevented the onset of septic shock (as determined by a sig- MAP Kinase = Mitogen Associated Protein Kinase
nificant fall in arterial blood pressure), ARDS (as determined MCP-1 = Monocyte Chemoattractant Protein-1
by a fall in PaO2/FiO2 ratio and an increase in lung water
(expressed as a wet to dried tissue ratio) and decreased se- MMP = Matrix Metalloproteinase
NE = Neutrophil Elastase
1
Salvatore, C.M.; Katz-Gaynor, K.; McCracken, R.D. and Hardy, R.D. Meeting Ab- NO = Nitric Oxide
stract B-401, ICAAC Conference, San Francisco 2006.
236 Endocrine, Metabolic & Immune Disorders - Drug Targets, 2007, Vol. 7, No. 3 Rempe et al.

NOS = Nitric Oxide Synthase [25] Gonzales, A.A.; Segura, A.M.; Horiba, K.; Qian, S.; Yu, Z.X.;
Stetler-Stevenson, W.; Willerson, J.T.; McAllister, H.A. Jr. and
TDI = Toluene Diisocyanate-Induced Ferrans, V.J. (2002) Hum. Pathol., 33 (12), 1158-1164.
[26] Garcia, R.A.; Pantazatos, D.P.; Gessner, C.R.; Go, K.V.; Woods,
REFERENCES V.L. Jr. and Villareal, F.J. (2005) Mol. Pharmacol., 67 (4), 1128-
1136.
[1] Stephens, C.R.; Conover, L.H.; Pasternack, R.; Hochstein, F.A.; [27] Lee, K.S.; Jin, S.M.; Kim, S.S. and Lee, Y.C. (2004) J. Allergy
Moreland, W.T.; Regna, P.P.; Pilgrim, F.J.; Brunnings, K.J. and Clin. Immunol., 113 (5), 902-909.
Woodward, R.B. (1954) J. Am. Chem. Soc., 76 (13), 3568-3575. [28] Palei, A.C.T.; Zaneti R.A.G.; Fortuna, G.M.; Gerlach, R.F. and
[2] Nelson, M.L. (1998) Adv. Dental Res., 12 (2), 5-11. Tanus-Santos, J.E. (2005) Angiology, 56 (5), 611-617.
[3] Sapadin, A.N. and Fleischmajer, R. (2006) J. Am. Acad. Dermatol., [29] Van den Steen, P.E.; Dubois, B.; Nelissen I.; Rudd, P.M.; Dwek,
54 (2), 258-265. R.A. and Opdenakker, G. (2002) Crit. Rev. Biochem. Mol. Biol., 37
[4] Golub, L.M.; Lee, H.M.; Lehrer, G.; Nemiroff, A.; McNamara, (6), 375-536.
T.F.; Kaplan, R. and Ramamurthy, N.S. (1983) J. Periodontol. [30] Battistini, B. (2003) Can. J. Physiol. Pharmacol., 81 (6), 555-569.
Res., 18 (5), 516-526. [31] Sepper, R.; Konttinen, Y.T.; Ding, Y.; Takagi, M. and Sorsa, T.
[5] Preshaw, P.M.; Hefti, A.F.; Jepsen, S.; Etienne, D.; Walker, C. and (1995) Chest, 107 (6), 1641-1647.
Bradshaw, M.H. (2004) J. Clin. Periodontol. Res., 31 (9), 697-707. [32] Guignabert, C.; Taysse, L.; Calvet, J.-H.; Planus, E.; Delamanche,
[6] Bikowski, J.B. (2003) Skinmed., 2 (4), 234-245. S.; Galiacy, S. and d’Ortho, M.-P. (2005) Am. J. Physiol. Lung Cell
[7] O’Dell, J.R.; Elliott, J.R.; Mallek, J.A.; Mikuls, T.R.; Weaver, Mol. Physiol., 289 (1), L67-L74.
C.A.; Glickstein, S.; Blakely, K.M.; Hausch, R. and Leff, R.D. [33] Rappeneau, S.; Baeza-Squiban, A.; Marano, F. and Calvet, J.-H.
(2006) Arthritis Rheum., 54 (2), 621-627. (2000) Toxicol. Sci., 58 (1), 153-160.
[8] Lauhio, A.; Sorsa, T.; Lindy, O.; Suomalainen, K.; Saari, H.; [34] Zeidler, P.; Hubbs, A.; Battlelli, L. and Castranova, V. (2004) J.
Golub, L.M. and Konttinen, Y.T. (1992) Arthritis Rheum., 35 (2), Toxicol. Environ. Health A, 67 (13), 1001-1026.
195-198. [35] Genovese, T.; Cuzzocrea, S.; Di Paola, R.; Failla, M.; Mazzon, E.;
[9] Stirling, D.P.; Koochesfahani, K.M.; Steeves, J.D. and Tetzlaff, W. Sortino, M.A.; Frasca, G.; Gili, E.; Crimi, N.; Caputi, A.P. and
(2005) Neuroscientist, 11 (4), 308-322. Vancheri, C. (2005) Respir. Res., 6, 58-74.
[10] Zemke, D. and Majid, A. (2004) Clin. Neuropharmacol., 27 (6), [36] Hoyt, J.C.; Ballering, J.; Numanami, H.; Hayden, J.M. and Rob-
293-298. bins, R.A. (2006) J. Immunol., 176 (1), 567-572.
[11] Zink, M.C.; Uhrlaub, J.; DeWitt, J.; Voelker, T.; Bullock, B.; [37] D’Agostino, P.; La Rosa, M.; Barbera, C.; Arcoleo, F.; Di Bella,
Mankowski, J.; Tarwater, P.; Clements, J. and Barber, S. (2005) J. G.; Milano, S. and Cillari, E. (1998) J. Infect. Dis., 177 (2), 489-
Am. Med. Assoc., 293 (16), 2003-2011. 492.
[12] Ingbar, D.H. (1995) in Pulmonary and Critical Care Medicine, Vol. [38] Shapiro, S.D.; Goldstein, N.M.; Houghton, A.M.; Kobayashi, D.K.;
1, (Bone, R.C.; Dantzker, D.R.; George, R.B.; Matthay, R.A. and Kelly, D. and Belaaouaj, A. (2003) Am. J. Pathol., 163 (6), 2329-
Reynolds, H.Y., Eds.) Mosby, St. Louis, MO, pp. A1-19. 2335.
[13] Valcke, Y.; Pauwels, R. and Van der Straeten, M. (1990) Eur. [39] Lee, C.T.; Fein, A.M.; Lippmann, M.; Holtzman, H.; Kimbel, P.
Respir. J., 3 (6), 715-722. and Weinbaum, G. (1981) N. Eng. J. Med., 304 (4), 192-196.
[14] Gartmann, J. (1975) Chemotherapy, 21 (Suppl. 1), 19-26. [40] O’Connor, C.M.; Gaffney, K.; Keane, J.; Southey A.; Bryne, N.;
[15] Hartnett, B.J.S. and Marlin, G.E. (1976) Thorax, 31 (2), 144-148. O’Mahoney, S. and Fitzgerald, M.X. (1993) Am. Rev. Respir. Dis.,
[16] Stockley, R.A. (1999) Am. J. Respir. Crit. Care Med., 160 (5 Pt 2), 148 (6 Pt 1), 1665-1670.
S49-S52. [41] Birrer, P.; McElvaney, N.G.; Rudeberg, A.; Sommer, C.W.;
[17] Nguyen, M.; Arkell, J. and Jackson, C.J. (2000) J. Biol. Chem., 275 Liechti-Gallati, S.; Kraemer, R.; Hubbard, R. and Crystal, R.G.
(13), 9095-9098. (1994) Am. J. Respir. Crit. Care Med., 150 (1) 207-213.
[18] Wilhelm, S.M.; Collier, I.E.; Marmer, B.L.; Eisen, A.Z.; Grant, [42] Mallya, S.K.; Hall, J.E.; Lee, H.M.; Roemer, E.J.; Simon, S.R. and
G.A. and Goldberg, G.I. (1989) J. Biol. Chem., 264 (29), 17213- Golub, L.M. (1994) Ann. N.Y. Acad. Sci., 732, 303-314.
17221. [43] Raza, M.; Ballering, J.G.; Hayden, J.M.; Robbins, R.A. and Hoyt,
[19] Ogata, Y.; Enghild, J.J. and Nagase, H. (1992) J. Biol. Chem., 267 J.C. (2006) Exp. Lung Res., 32 (1-2), 15-26.
(6), 3581-3584. [44] Golub, L.M.; McNamara, T.F.; D’Angelo, G.; Greenwald, R.A.
[20] Delclaux, C.; d’Ortho, M.P.; Delacourt, C.; Lebargy, F.; Brun- and Ramamurthy, N.S. (1987) J. Dent. Res., 66 (8), 1310-1314.
Buisson, C.; Brochard, L.; Lemaire, F.; Lafuma, C. and Harf, A. [45] Tolomeo, M.; Grimaudo, S.; Milano, S.; La Rosa, M.; Ferlazzo, V.;
(1997) Lung Cell Mol. Physiol., 272 (3), L442-L451. Di Bella, G.; Barbera, C.; Simoni, D.; D’Agostino, P. and Cillari,
[21] Torii, K.; Iida, K.; Miyazaki, Y.; Saga, S.; Kondoh, Y.; Taniguchi, E. (2001) Br. J. Pharmacol., 133 (2), 306-314.
H.; Taki, F.; Takagi, K.; Matsuyama, M. and Suzuki, R. (1997) Am. [46] Steinberg, J.; Halter, J.; Schiller, H.; Gatto, L.; Carney, D.; Lee, H.-
J. Respir. Crit. Care Med., 155 (1), 43-46. M.; Golub, L. and Nieman, G. (2005) Shock, 24 (4), 348-356.
[22] Sagel, S.D.; Kapsner, R.K. and Osberg, I. (2005) Pediatr. Pul- [47] Carney, D.E.; McCann, U.G.; Schiller, H.J.; Gatto, L.A.; Steinberg, J.;
monol., 39 (3), 224-232. Picone, A.L. and Nieman, G.F. (2001) J. Surg. Res., 99 (2), 245-252.
[23] Ratjen, F.; Hartog, C.M.; Paul, K.; Wermelt, J. and Braun, J. (2002) [48] Liu, Y.; Ryan, M.E.; Lee, H.-M.; Simon, S.; Tortora, G.; Lauzon,
Thorax., 57 (11), 930-934. C.; Leung, M.K. and Golub, L.M. (2002) Antimicrob. Agents Che-
[24] Hartog, C.M.; Wermelt, J.A.; Sommerfeld, C.O.; Eichler, W.; mother., 46 (5), 1447-1454.
Dalhoff, K. and Braun, J. (2003) Am. J. Respir. Crit. Care Med., [49] Liu, Y.; Tortora, G.; Ryan, M.E.; Lee, H.-M. and Golub, L.M.
167 (4), 593-598. (2002) Antimicrob. Agents Chemother., 46 (5), 1455-1461.

Received: 31 October, 2006 Accepted: 13 December, 2006

You might also like