ArticlePDF Available

New Insights Into the Pathogenesis of Cystic Fibrosis

Authors:

Abstract and Figures

The cystic fibrosis transmembrane regulator (CFTR) should no longer be viewed primarily as a ‘chloride channel’ but recognized as a channel that also controls the efflux of other physiologically important anions, such as glutathione (GSH) and bicarbonate. More effective approaches to cystic fibrosis treatment may result from this reconceptualization of the CFTR by researchers and clinicians. For example, oxidant damage in cystic fibrosis has been assumed to be a significant part of the pathophysiology of the disease. Generally speaking, antioxidant status in cystic fibrosis is compromised. However, until recently this was seen as secondary to the excessive chemoattraction of neutrophils in this disease caused by mutation of the CFTR protein, leading to a high oxidant burden. New findings suggest that the cystic fibrosis mutations in fact cause a primary dysfunction in the system of one of the body’s most important antioxidant and immune-signaling substances: the reduced GSH system. Cystic fibrosis mutations significantly decrease GSH efflux from cells without redundant channels to the CFTR; this leads to deficiency of GSH in the epithelial lining fluid of the lung, as well as in other compartments, including immune system cells and the gastrointestinal tract. This deficiency is exaggerated over time as the higher-than-normal oxidant burden of cystic fibrosis leads to successively larger decrements in GSH without the normal opportunity to fully recover physiologic levels. This GSH system dysfunction may be the trigger for initial depletion of other antioxidants and may also play a role in initiating the over-inflammation characteristic of cystic fibrosis. Proper GSH system functioning also affects immune system competence and mucus viscosity, both of relevance to cystic fibrosis pathophysiology. In a way, cystic fibrosis may be thought of as the first identified disease with GSH system dysfunction. This overview provides a review of the most pertinent recent research findings in this area. Exogenous augmentation of GSH in the lung epithelial lining fluid is possible, and therapeutic approaches include administration of aerosolized buffered GSH, intravenous GSH, and oral GSH. However, it is important to remember that the pathophysiology of cystic fibrosis is multifactorial, and rectification of GSH system dysfunction in patients with cystic fibrosis will not eliminate all harmful effects of the disease. The promising results of two clinical trials of aerosolized buffered GSH in cystic fibrosis patients have been published or accepted for publication at the time of this writing. GSH depletion in lung epithelial lining fluid has also been noted in other respiratory diseases such as COPD, idiopathic pulmonary fibrosis, and adult respiratory distress syndrome, and therapies to augment GSH may also be contemplated in these diseases.
Content may be subject to copyright.
This material is
the copyright of the
original publisher.
Unauthorised copying
and distribution
is prohibited.
Treat Respir Med 2004; 3 (6): 353-363
L
EADING
A
RTICLE
1176-3450/04/0006-0353/$31.00/0
2004 Adis Data Information BV. All rights reserved.
New Insights Into the Pathogenesis of
Cystic Fibrosis
Pivotal Role of Glutathione System Dysfunction and Implications for Therapy
Valerie M. Hudson
Brigham Young University, Provo, Utah, USA
The cystic fibrosis transmembrane regulator (CFTR) should no longer be viewed primarily as a ‘chloride
Abstract
channel’ but recognized as a channel that also controls the efflux of other physiologically important anions, such
as glutathione (GSH) and bicarbonate. More effective approaches to cystic fibrosis treatment may result from
this reconceptualization of the CFTR by researchers and clinicians. For example, oxidant damage in cystic
fibrosis has been assumed to be a significant part of the pathophysiology of the disease. Generally speaking,
antioxidant status in cystic fibrosis is compromised. However, until recently this was seen as secondary to the
excessive chemoattraction of neutrophils in this disease caused by mutation of the CFTR protein, leading to a
high oxidant burden. New findings suggest that the cystic fibrosis mutations in fact cause a primary dysfunction
in the system of one of the body’s most important antioxidant and immune-signaling substances: the reduced
GSH system. Cystic fibrosis mutations significantly decrease GSH efflux from cells without redundant channels
to the CFTR; this leads to deficiency of GSH in the epithelial lining fluid of the lung, as well as in other
compartments, including immune system cells and the gastrointestinal tract. This deficiency is exaggerated over
time as the higher-than-normal oxidant burden of cystic fibrosis leads to successively larger decrements in GSH
without the normal opportunity to fully recover physiologic levels. This GSH system dysfunction may be the
trigger for initial depletion of other antioxidants and may also play a role in initiating the over-inflammation
characteristic of cystic fibrosis. Proper GSH system functioning also affects immune system competence and
mucus viscosity, both of relevance to cystic fibrosis pathophysiology. In a way, cystic fibrosis may be thought of
as the first identified disease with GSH system dysfunction.
This overview provides a review of the most pertinent recent research findings in this area. Exogenous
augmentation of GSH in the lung epithelial lining fluid is possible, and therapeutic approaches include
administration of aerosolized buffered GSH, intravenous GSH, and oral GSH. However, it is important to
remember that the pathophysiology of cystic fibrosis is multifactorial, and rectification of GSH system
dysfunction in patients with cystic fibrosis will not eliminate all harmful effects of the disease. The promising
results of two clinical trials of aerosolized buffered GSH in cystic fibrosis patients have been published or
accepted for publication at the time of this writing. GSH depletion in lung epithelial lining fluid has also been
noted in other respiratory diseases such as COPD, idiopathic pulmonary fibrosis, and adult respiratory distress
syndrome, and therapies to augment GSH may also be contemplated in these diseases.
Cystic fibrosis disease is the result of a mutation of the cystic colonization of the lungs by bacteria and fungi and auto-destruc-
fibrosis transmembrane regulator (CFTR) protein, resulting in tion of lung tissue by excessive inflammation even in the absence
missing or defective cellular anion efflux channels in epithelial of pathogen challenge (and accelerated by pathogen challenge
cells. Most cystic fibrosis patients ultimately die of respiratory when it occurs). The excessive inflammation has been linked to
failure, as a result of deterioration in pulmonary function. There the cystic fibrosis mutation itself and is associated with greater
are several causes for deterioration in lung function, including than normal chemoattraction for neutrophils as a result of higher
This material is
the copyright of the
original publisher.
Unauthorised copying
and distribution
is prohibited.
354 Hudson
constitutive levels of, for example, interleukin (IL)-8.[1-8] The important roles of GSH, then, is to act as a water-soluble antioxi-
oxidant burden caused by such excessive inflammation appears to dant. Not only can it neutralize oxidants through an enzymatic
overwhelm natural antioxidant defenses. The resulting damage to pathway utilizing GSH peroxidase, it is also capable of neutraliz-
lung tissue allows for greater adhesion of pathogens,[9] in addition ing oxidants directly without the use of an enzymatic pathway.[16]
to reducing lung function directly.[10] In cystic fibrosis, however, epithelial cells still produce GSH
New research findings, however, suggest that the traditional normally, but one result is significant impairment of the ability of
view of disease pathogenesis needs some modification. It now cells that do not possess a channel redundant to the CFTR to efflux
appears that cystic fibrosis mutations cause a primary dysfunction GSH to fulfill its functions in the extracellular milieu.
in one of the most important antioxidant and immune-signaling Each antioxidant system – fat-soluble, water-soluble, and enzy-
substances: the reduced glutathione (GSH) system. Dysfunction in matic – protects the cell within its own sphere of action. Some
the GSH system may be the catalyst for initial depletion of other systems operate within the cytosol, which others operate at the cell
antioxidants and may play a role in priming and perpetuating membrane or are active in the extracellular milieu. GSH operates
excessive inflammation characteristic of cystic fibrosis. Neverthe- both within the cytosol and in the extracellular milieu. In these
less, it is important to remember that cystic fibrosis pathophysiolo- compartments, it is capable of directly reducing oxidants and it
gy is multifactorial and that not all cystic fibrosis disease manifes- also reacts with GSH peroxidase, located in the cell membrane, to
tations can be linked to GSH system dysfunction. neutralize oxidants. GSH is replenished in two ways: by interac-
This modification of the traditional view is in line with tion of GSH disulfide (GSSG) with the enzyme GSH reductase
cutting-edge cystic fibrosis research, which suggests that the sole and by synthesis of GSH within the cell (i.e. de novo or after
focus on the CFTR channel as a chloride efflux channel has cleavage of extracellular GSH and transport of component amino
obscured other significant functions associated with it. Current acids back into the cell). Circulation of GSH effluxed from cells
research now views the CFTR channel as, at the very least, a throughout the body may allow for higher levels of GSH particu-
chloride, bicarbonate, and GSH efflux channel. Undoubtedly this larly in the extracellular compartments, such as the lung.
list will be expanded in the future. This new and fundamental shift The various antioxidant systems are interdependent, to one
in the way the CFTR channel is viewed may allow for the degree or another, for proper function. The crippling of one system
development of innovative and effective therapies for cystic fibro- leads to decreased protection by other antioxidant systems. With-
sis. In this article, we will focus on the CFTR as a GSH efflux out GSH, as we have seen, GSH peroxidase is unable to function
channel and discuss its significance for the respiratory system. It is as an antioxidant.[17] Antioxidant systems such as GSH, ascorbic
worth noting that preliminary studies of the role of bicarbonate acid, tocopherol, and ubiquinol-10[18,19] are interdependent, and
secretion in the lung have shown that the lack of functional CFTR normal levels of each in reduced form are required to maintain
may result in a decrease in the pH of the cystic fibrosis lung, which
normal levels of the others in reduced form.[20] In addition, GSH
may also have pathologic consequences;[11,12] however, this find-
deficiency is linked to decreased activity of catalase and superox-
ing is disputed elsewhere.[13-15] The newly understood role of
ide dismutase.[20] GSH deficiency also taxes the fat-soluble antiox-
CFTR as a GSH and bicarbonate efflux channel also has important
idant systems by permitting greater levels of lipid peroxidation,
implications for gastrointestinal complications associated with
yielding damaging metabolites.[20-27] This genetic chink in the
cystic fibrosis. In this article, discussion will be confined to the
antioxidant armor of cystic fibrosis patients predisposes them to
effects of CFTR mutation on the respiratory system only.
have successively larger decrements in antioxidant protection over
time, as other antioxidants are consumed in greater quantities or
1. Understanding the Role of the Glutathione (GSH) left unused as a result of impaired GSH efflux.
System in Normal Lung Health
Furthermore, recent studies have demonstrated the importance
of S-glutathiolation of proteins under conditions of oxidative or
Virtually all cells of the body produce thiol-reduced GSH from
nitrosative stress.[28,29] To prevent irreversible loss of intracellular
the three amino acids glutamine, glycine, and cysteine. Cysteine
and extracellular protein function under such stress, mixed disul-
serves as the rate-limiting amino acid for GSH production. It is
fides are formed between protein cysteines and GSH. These S-
estimated that an adult male produces approximately 10g of GSH
glutathiolated proteins are more stable and can be dethiolated by
per day. Not only is GSH present in the cells of the body, it also
either non-enzymatic reduction or enzymatic cleavage of the disul-
bathes the extracellular spaces of the body, with high extracellular
fide bond. Thus, S-glutathiolation allows for reversible regulation,
levels in organ systems that come in contact with the oxidant-rich
and therefore generalized protection, of sensitive proteins.
atmosphere, such as the cornea and the lung. One of the most
2004 Adis Data Information BV. All rights reserved. Treat Respir Med 2004; 3 (6)
This material is
the copyright of the
original publisher.
Unauthorised copying
and distribution
is prohibited.
Pathogenesis of Cystic Fibrosis 355
A disrupted systemic antioxidant shield leads to predictable total GSH within such cells may remain normal, but the
damage to lung tissue by oxidants. Oxidants directly harm sensi- GSH :GSSG ratio may become substantially decreased. Howev-
tive lung epithelia. In addition, they are able to inactivate er, immune system cells are among the class of cells that have
antiproteases, which then leads to increased elastase damage, channels redundant to the CFTR. With a growing extracellular
increased mucus secretion, and deranged immune signaling.[30-34] deficit, immune system cells may actually attempt to efflux GSH
Oxidants also adversely affect ciliary beat function in the lung, and to rectify that deficit. Furthermore, with the increasing oxidant
lung surfactant levels are diminished by the oxidant burden.[33] burden, immune system cells may use up their stores of GSH in
There is an increased production of chloramines, which further self-protection. Immune cells then become GSH deficient. All in
decreases epithelial integrity.[35-37] A higher oxidant burden also all, what the body senses in cystic fibrosis is that there is some
creates cell structure abnormalities, which may lead to impaired threat that is using up all of the GSH in oxidative reactions, even
cell function or even premature cell death.[38-53] In the lung, though that threat is nonexistent. What is really occurring in cystic
generalized bronchoconstriction can be another consequence of fibrosis is defective GSH efflux, but the body has no way of telling
decreased antioxidant functioning.[54] Damage to the epithelial the difference.
tissue of the lung also permits greater adhesion of pathogens.[55] The body responds by mobilizing itself to meet the nonexistent
Oxidants can also inactivate other parts of the GSH system, such threat. In short, it inflames. GSH deficiency in leukocytes causes
as GSH reductase and γ-glutamylcysteine transferase, both neces- increased release of oxidants such as hydrogen peroxide.[70] Cellu-
sary for cellular protection and proper redox functionality.[56-58] lar GSH deficiency causes increased transcription of nuclear fac-
Protein S-glutathiolation will decrease, resulting in irreversible tor-κB, which then codes for greater levels of inflammatory
loss of sensitive protein function.[29] cytokines, such as tumor necrosis factor-α, activator protein-1,
A second consequence of impaired GSH efflux is increased monocyte chemoattractant protein-1, IL-8, and IL-1a.[71-85] Such a
viscosity of mucus. GSH plays an important role in mucolysis of cytokine profile creates inflammation and recruitment of neutro-
disulfide bonds in mucus, in much the same manner as the more phils and macrophages even in the absence of a threat, which is
well known cysteine donors such as N-acetylcysteine (NAC).[59] precisely what occurs in cystic fibrosis. (Of course, when a patho-
Increased viscosity of mucus has important consequences to the gen threat does present itself, the inflammation becomes even
lung environment.[59-67] more excessive.) As long as full GSH replenishment cannot occur
(because of defective GSH efflux from most of the cells of the
Finally, the redox system of GSH, as indicated by the
body), the inflammation will continue and become chronic, as it is
GSH :GSSG ratio manifesting redox potential, is an important
in cystic fibrosis.
immune system signal. The GSH :GSSG ratio is usually greater
than 9 :1, sometimes reaching over 100–200 :1, depending on the In addition to chronic inflammation, the continuing inability to
compartment. When that ratio is substantially decreased or there is replenish GSH, especially in immune cells, creates a situation of
a decrease in total GSH (GSH + GSSG), the body appears to immune incompetence. GSH deficiency in leukocytes causes, in
interpret such events as a call for assistance from the immune general, impaired release of lysosomal enzymes, decreased phago-
system to cope with some threatening challenge that is resulting in cytosis, and premature apoptosis.[70,86-100] GSH deficiency also
pathologic oxidative reactions that are outpacing GSH replenish- creates a situation of incomplete immune system signaling, be-
ment. For example, Day and colleagues[68,69] have found that when cause GSH reduction of disulfide bonds is necessary for such
normal mouse lung tissue is challenged with Pseudomonas aerugi- signaling. For example, antigen-presenting cells use the reductive
nosa, there is a 3-fold induction in epithelial lining fluid (ELF) power of GSH to present antigens to T cells.[101-104] B cells appear
GSH levels and a 2-fold induction in CFTR levels, presumably to similarly affected,[105,106] and activation of T and B cells appears
offset increased oxidative reactions. The inability to effect this related to GSH levels.[107-112] Interferon-γ signaling is also depen-
large increase in ELF GSH because of ineffectual transport due to dent on the presence of GSH.[113] Such interruptions of appropriate
CFTR mutation will substantially alter both the redox ratio and the immune signaling begin to shift the organism to a more T helper-2
level of total GSH. type of response, which is less effective in pathogen clear-
It is important to understand how the body reads the effects of ing.[101,107,114-118] In addition, GSH is necessary to create a reservoir
CFTR absence or malfunction on the GSH system. Cells without of nitric oxide (NO) [via creation of s-nitrosoglutathione
channels redundant to CFTR, such as lung epithelia (whose chan- (GSNO)], and the lack of such a reservoir leads to a generalized
nels redundant to the CFTR are at the basolateral, not the apical, lack of NO itself in the lung environment.[119-130] NO not only has
surface), will not be able to export GSH to the extracellular milieu, important bactericidal properties but also is necessary in cell
and the extracellular deficit may become quite severe. Levels of signaling and smooth muscle relaxation and helps regulate ciliary
2004 Adis Data Information BV. All rights reserved. Treat Respir Med 2004; 3 (6)
This material is
the copyright of the
original publisher.
Unauthorised copying
and distribution
is prohibited.
356 Hudson
beat function.[131-137] When antioxidant defenses are compromised, 2.1 Effect of Cystic Fibrosis Transmembrane Regulator
superoxide anion will scavenge NO almost instantaneously, the Mutation on Efflux of GSH
reaction limited only by the extent of diffusion. The depletion of
NO has important consequences for both lung function and im- In retrospect, it was the work of Linsdell and Hanrahan[138] that
mune function. first identified that the CFTR channel played a role in the efflux of
In short, then, a generalized GSH deficiency will cause inflam- GSH. Afer clamping the CFTR channels of Chinese hamster ovary
mation coupled, paradoxically, with decreased immune system cells, Linsdell and Hanrahan[138] compiled a list of substances that
competence to clear pathogens. These effects are in addition to the were subsequently not effluxed. GSH was on the list. Since that
loss of antioxidant protection and mucolytic activity noted above study published in 1998, however, it was the work of Gao and
in connection with GSH deficiency. The CFTR mutations that colleagues[139] that arguably pushed that insight further. Using cell
cause cystic fibrosis produce these consequences as a result of lines of cystic fibrosis lung epithelia, this team was able to
severely impaired efflux of GSH from most cells of the body. demonstrate markedly decreased GSH efflux. Velsor and col-
Figure 1 represents a summary of how diminished GSH transport leagues[140] and Day and colleagues[69] also found a 50% reduction
influences the pathophysiology of cystic fibrosis. in GSH levels in the ELF of the lung in uninfected CFTR knockout
mice and a lack of normal induction of GSH in the ELF when
2. GSH System Dysfunction in Cystic Fibrosis challenged by P. aeruginosa. Kogan and colleagues[141,142] found
The evidence for a primary GSH system dysfunction in cystic this same diminished GSH efflux with a variety of CFTR mutants,
fibrosis is steadily growing. including G551D, R347D, K464A, and K1250A, and through the
use of sophisticated tests were able to confirm that purified CFTR
protein alone directly mediated nucleotide-dependent GSH flux
and not via other associated chloride transport proteins.[142] Finder
et al.[143] also document a 49% reduction of GSH in bronchoalveo-
lar lavage fluid from cystic patients with fibrosis. The findings
Oxidant injury is unchecked, and
chronic inflammation begins
Leukocytes are drawn into the airways, and
GSH leaks out of the PMNs through MRP
Mucous thickens
Leukocytes are then depleted of intracellular GSH
necessary for phagocytosis,
oxidative burst, and other normal function.
This leads to premature apoptosis
Weakened immunity results in
colonization of bacteria
Chronic infection and inflammation results in increased
elastase activity and destruction of lung structures;
bronchiectesis develops
S-Glutathiolation
impaired
GSH-depleted
leukocytes transcribe
NF-κB, amplifying
inflammatory cascade
Preserved epithelial
intracellular GSH
impairs apoptosis
of infected lung
epithelium
Other antioxidants
are depleted
Extracellularly, total GSH and GSH/GSSG ratios are abnormally low
Defective CFTR protein in membrane does not allow
normal egress of GSH from lung epithelial cells
Fig. 1. The influence of diminished glutathione transport on the pathophysiology of cystic fibrosis. CFTR = cystic fibrosis transmembrane regulator; GSH =
glutathione; GSSG = glutathione disulfide; MRP = multidrug resistance-associated protein; NF-κB = nuclear transcription factor κB; PMN =
pol
y
morphonuclear leukoc
y
te.
2004 Adis Data Information BV. All rights reserved. Treat Respir Med 2004; 3 (6)
This material is
the copyright of the
original publisher.
Unauthorised copying
and distribution
is prohibited.
Pathogenesis of Cystic Fibrosis 357
from these five research teams constitute the ‘smoking gun’: cystic found substantially lower levels of GSH in sputum neutrophils
fibrosis directly causes significantly impaired GSH efflux. from patients with cystic fibrosis compared with blood neutro-
phils, which also correlated with cell death rates. Furthermore, in
this compartment, alterations in the expression of channels redun-
2.2 GSH Deficiency in the Cystic Fibrosis dant to the CFTR have also been noted: for example, multidrug
Lung Environment resistance-associated protein 1 has been shown to be upregulated
in cystic fibrosis erythrocytes.[150]
If we conceive of three distinct compartments in the cystic
fibrosis lung environment, we find empirical evidence that GSH 2.3 Related Phenomena
deficiency arises in the two compartments where the deficiency
would be expected if a transport abnormality were at fault. The As part of this hypothesis of mutation-derived GSH efflux
three compartments to visualize are: (i) cells that do not have a impairment, we should find that, other than the effects of the
channel redundant to the CFTR (at least at the apical surface), such mutation, the rest of the GSH antioxidant system remains intact in
as the epithelium of the lung; (ii) the extracellular milieu, com- patients with cystic fibrosis. This is in fact the case: at least normal
posed primarily of the lung ELF; and (iii) cells that do have levels of GSH peroxidase and γ-glutamylcysteine synthetase and
channels redundant to the CFTR (such as leukocytes or erythro- increased levels of γ-glutamylcysteine transferase, γ-glutamyl
cytes). transpeptidase, and GSH reductase have been found in patients
Gao and colleagues[139] found normal levels of total GSH in the with cystic fibrosis.[17,144,148,151-153] Therefore, the observed GSH
first compartment, which included cystic fibrosis lung epithelial deficiency is a primary deficiency not caused by malfunctions in
cells that did not have a channel redundant to the CFTR (at least at other parts of the overall GSH antioxidant system.
the apical surface). This should be expected, as there is no GSH
synthesis defect in cystic fibrosis as there is, say, in AIDS. The 2.4 Impaired GSH Efflux and Cystic Fibrosis Pathology
GSH :GSSG ratio in these cells was not ascertained and remains
to be analyzed. Note that the lack of ability to be depleted of GSH Many of the effects that would be expected from impaired GSH
may lead to decreased levels of appropriate apoptosis when these efflux in most cells have been noted as part of cystic fibrosis
cells are infiltrated by pathogens. pathology.
In the second compartment, consisting of ELF, several studies Inflammation in the absence of pathogen challenge has been
have found progressive GSH deficiency arising and persisting noted in the youngest of infants with cystic fibrosis.[1,2,154-157]
over time in patients with cystic fibrosis. Hull et al.[144] found that Other antioxidant systems of the body have been found to be
non-infected cystic fibrosis do not appear to have a GSH deficien- compromised in cystic fibrosis. This is in part a result of fat
cy in their ELF, though only the total GSH, and not the malabsorption due to pancreatic insufficiency in most patients
GSH :GSSG ratio, was analyzed. An altered GSH :GSSG ratio in with cystic fibrosis, resulting in lower levels of retinol and
the ELF of infants with cystic fibrosis would be evidence that the tocopherol, but this situation can be accelerated and aggravated by
impaired GSH efflux has begun to have an impact in the respirato- GSH deficiency. Affected systems include at least retinol, beta-
ry system of these patients.[145,146] Hull et al.[144] did find that carotene, tocopherol, activity of GSH peroxidase, ascorbic acid,
infected infants with cystic fibrosis infants had slightly lower total and activity of superoxide dismutase.[17,158-163] Antiproteases have
levels of GSH in their ELF. Brown et al.[147] determined that, been shown to be neutralized in cystic fibrosis, and surfactant
beyond infancy, plasma sulfhydryls decreased significantly with levels are lower.[164,165] There is an altered cytokine profile conso-
age in patients with cystic fibrosis. Roum et al.[148] found a nant with GSH deficiency, and also exhaled NO is not elevated, as
profound deficiency of GSH in the ELF in adult patients with it is in other respiratory diseases with a high oxidant bur-
cystic fibrosis, with levels of 5–10% of normal when oxidant den.[1-8,166-178] Interestingly, in vitro, the addition of S-nitrosogluta-
burden was factored in. This team also found plasma GSH levels thione to delF508 cystic fibrosis cell lines has been shown to help
of about 50% of normal in these adult patients with cystic fibrosis; in the maturation and functionality of the mutated protein.[179,180]
in both the ELF and plasma, they also found an extremely de- Furthermore, decreased apoptosis has been noted in pathogen-
creased GSH :GSSG ratio. Finder et al.[143] found a 49% reduction infiltrated cells without redundant anion channels in cystic fibro-
of GSH in bronchoalveolar lavage fluid from patients with cystic sis.[181]
fibrosis. In conclusion, the view that CFTR acts as an important GSH
In the third compartment, including cells with channels redun- efflux channel is gaining strength through recent empirical re-
dant to the CFTR, in the lung environment, Tirouvanziam et al.[149] search findings. In addition to several ‘smoking guns’, the related
2004 Adis Data Information BV. All rights reserved. Treat Respir Med 2004; 3 (6)
This material is
the copyright of the
original publisher.
Unauthorised copying
and distribution
is prohibited.
358 Hudson
phenomena and effects that one would expect if cystic fibrosis indicators, including peak flow, and in compliance analysis,
caused GSH efflux impairment are also empirically demonstrable cough.
(figure 1). This evidence leads us to inquire about therapeutic The results of these two clinical trials of aerosolized buffered
implications. However, it is important to remember that cystic GSH are very promising and warrant larger, multicenter trials of
fibrosis pathophysiology is multifactorial, and rectification of longer duration.
GSH system dysfunction in patients with cystic fibrosis will not Oral administration of GSH is not to be overlooked. Previously,
eliminate all harmful effects of the disease. researchers could not agree on whether GSH was cleaved in the
digestive tract or taken up intact in the jejunum. Newer studies
seem to indicate the latter.[196-200] Furthermore, there is new and
3. Therapeutic Implications innovative research being conducted to create a novel peptide that
could serve as a GSH efflux for cystic fibrosis cells.[201] Finally,
The usual and most direct route to augment GSH levels is to intravenous GSH has been used as a treatment for radiation
provide a cysteine donor, such as NAC, to the patient. As cysteine poisoning, as well as for other diseases such as Parkinson dis-
is the rate-limiting amino acid for GSH synthesis, this route is ease.[183,202,203] Given that it is most likely that the lung is a net
generally effective in otherwise healthy individuals. However, as importer of circulating GSH, this route might bear further investi-
we have seen, GSH synthesis is not impaired in cystic fibrosis; the gation in the case of cystic fibrosis.
problem is in GSH efflux from the cells in which GSH is synthe- Other respiratory ailments are marked by a decrease in GSH in
sized. Nevertheless, Hosseini and colleagues[182] have used a cys- the ELF. Clinicians treating such illnesses may want to examine
teine-rich whey powder to treat C57B1/6 mice infected with GSH augmentation in diseases such as adult respiratory distress
Pseudomonas spp. and noted some improvement in mortality; syndrome (ARDS), COPD, idiopathic interstitial pneumonia, IPF
therefore, precursors might usefully complement a strategy of of nonsmokers, idiopathic respiratory distress syndrome, and dif-
exogenous GSH augmentation. fuse fibrosing alveolitis.
Direct augmentation of GSH levels in the ELF with aerosolized In summary, augmentation of GSH in the ELF is feasible and
GSH has been carried out in vivo, including in patients with cystic may be useful not only for cystic fibrosis, but also for several other
fibrosis, AIDS, idiopathic pulmonary fibrosis (IPF), COPD, and respiratory conditions. Given the vasodilatory and anti-inflam-
other diseases.[183-191] Unfortunately, GSH in solution has a pH of matory properties of GSH, there may be contraindications to its
2.7 and is an irritant to the lung. This has hampered the usefulness use. It could be speculated that in patients with a history of
of this therapy for patients with respiratory ailments. Two clinical hemoptysis/pneumothorax, those yielding a positive culture for
trials, using buffered GSH with a pH of 5–6, have been carried out Burkholderia cepacia, or those with an FEV1 <30% predicted, the
in patients with cystic fibrosis. use of GSH may be contraindicated until further, more extensive
In the first trial, using the AKITA1 inhalation device, Griese trials have been conducted.
et al.[192] were able to increase the GSH level in bronchoalveolar
lavage fluid in patients with cystic fibrosis through inhalation of a 4. Conclusion
buffered GSH solution. One hour after inhalation, GSH levels
increased 3- to 4-fold, and at 12 hours levels of GSH were still In conclusion, new research is beginning to alter our under-
almost double those at baseline. Griese et al. found that with 14 standing of the CFTR channel. It is no longer possible to view it
days’ use of three times daily buffered GSH 300–450mg, FEV1merely or even primarily as a chloride efflux channel. At this point
and FVC increased an average of 6–7% over baseline (p < in time, it must be viewed as a chloride/GSH/bicarbonate channel,
0.001).[193] No change in oxidative markers was observed, though though this list may grow in the future. As we more fully under-
this might be because of the short duration of therapy. stand the nature and functions of the CFTR channel, new therapeu-
tic approaches will begin to come into view, as we have seen with
The second study carried out by Bishop et al.[194,195] was a
GSH. In one respect, cystic fibrosis may be viewed (at least in
randomized, double-blind, placebo-controlled trial. The dosage of
part) as the first identified disease with GSH transport dysfunction.
buffered GSH was 66 mg/kg/day, divided into four inhalation
sessions over a 6-week period. Results indicated that 11 of 13 Clinicians may be able to make effective use of these new
clinical indicators examined favored the GSH treatment group insights from cutting-edge research. However, it is important to
over the placebo group, including lung function scores, and statis- remember that the pathophysiology of cystic fibrosis is multifacto-
tical significance was achieved in improvement in several of the rial, and rectification of GSH system dysfunction in cystic fibrosis
1The use of trade names is for product identification purposes only and does not imply endorsement.
2004 Adis Data Information BV. All rights reserved. Treat Respir Med 2004; 3 (6)
This material is
the copyright of the
original publisher.
Unauthorised copying
and distribution
is prohibited.
Pathogenesis of Cystic Fibrosis 359
15. Jayaraman S, Song Y, Verkman AS. Airway surface liquid pH regulation in well-
patients will not eliminate all harmful effects of the disease. differentiated airway epithelial cell cultures and mouse trachea. Am J Physiol
Indeed, some clinicians report that a significant proportion of their Cell Physiol 2001; 281 (5): C1504-11
16. Hudson VM. Rethinking cystic fibrosis pathology: the critical role of abnormal
patients with cystic fibrosis are already using GSH and/or NAC reduced glutathione (GSH) transport caused by CFTR mutation. Free Radic
without the physician’s knowledge.[204] Clinicians should inquire Biol Med 2001; 30 (12): 1440-61
from their patients with cystic fibrosis if they are already using 17. Benabdeslam H, Abidi H, Garcia I, et al. Lipid peroxidation and antioxidant
defenses in cystic fibrosis patients. Clin Chem Lab Med 1999 May; 37 (5): 511-
GSH/NAC, in order to monitor subsequent developments. In 6
addition, patients may be unaware of possible contraindications, 18. Stocker R, Bowry VW, Frei B, et al. Ubiquinol-10 protects human low density
the importance of pH adjustment for aerosol use, or the inherent lipoprotein more efficiently against lipid peroxidation than does alpha-tocoph-
erol. Proc Natl Acad Sci U S A 1991 Mar; 88: 1646-50
problems connected with NAC or other cysteine donor use alone. 19. Thomas SR, Neuzil J, Stocker R. Cosupplementation with coenzyme Q prevents
In short, given increasing patient interest in a growing body of the prooxidant effect of alpha-tocopherol and increases the resistance of LDL to
empirical evidence demonstrating mutation-derived GSH system transition metal-dependent oxidation initiation. Aterioscler Thromb Vasc Biol
1996 May; 16 (5): 687-96
dysfunction in cystic fibrosis, it behooves the clinician to learn as 20. Thanislass J, Raveendran M, Devaraj H. Buthionine sulfoximine-induced gluta-
much about this area of research as possible in order to treat cystic thione depletion: its effect on antioxidants, lipid peroxidation and calcium
homeostasis in the lung. Biochem Pharmacol 1995 Jul 17; 50 (2): 229-34
fibrosis patients in a more informed manner. Furthermore, clini-
21. Leedle RA, Aust SD. The effect of glutathione on the vitamin E requirement for
cians may find GSH augmentation a useful intervention for other inhibition of liver microsomal lipid peroxidation. Lipids 1990 May; 25 (5): 241-
respiratory diseases, as noted in section 3, such as COPD, ARDS, 5
and IPF. 22. Higuchi Y, Matsukawa S. Glutathione depletion induces giant DNA and high-
molecular-weight DNA fragmentation associated with apoptosis through lipid
peroxidation and protein kinase C activation in C6 glioma cells. Arch Biochem
Biphys 1999 Mar 1; 363 (1): 33-42
Acknowledgment 23. Scholz RW, Reddy PV, Wynn MK, et al. Glutathione-dependent factors and
inhibition of rat liver microsomal lipid peroxidation. Free Radic Biol Med
The authors have provided no information on sources of funding or 1997; 23 (5): 815-28
conflicts of interest directly relevant to the content of this review. 24. Hagiwara K, Naito K, Kurokawa Y, et al. Kidney injury induced by lipid peroxide
produced by vitamin E deficiency and GSH depletion in rats. J Nutr Sci
Vitaminol (Tokyo) 1991 Feb; 37 (1): 99-107
25. Angelini P, Cirelli F, Quarticelli A, et al. Administration of glutathione and lipid
References peroxidation induced during fasting. Boll Soc Ital Biol Sper 1990 Nov; 66 (11):
1. Khan TZ, Wagener JS, Bost T, et al. Early pulmonary inflammation in infants with 1097-104
cystic fibrosis. Am J Respir Crit Care Med 1995 Apr; 151 (4): 1075-82
26. Rosenblat M, Aviram M. Macrophage glutathione content and glutathione peroxid-
2. Balough K, McCubbin M, Weinberger M, et al. The relationship between infection
ase activity are inversely related to cell-mediated oxidation of LDL: in vitro and
and inflammation in the early stages of lung disease from cystic fibrosis. Pediatr
in vivo studies. Free Radic Biol Med 1998 Jan 15; 24 (2): 305-17
Pulmonol 1995 Aug; 20 (2): 63-70
3. Muhlebach MS, Stewart PW, Leigh MW, et al. Quantitation of inflammatory 27. Chen Q, Galleano M, Cederbaum AI. Cytotoxicity and apoptosis produced by
responses to bacteria in young cystic fibrosis and control patients. Am J Respir arachidonic acid in Hep G2 cells overexpressing human cytochrome P4502E1.
Crit Care Med 1999 Jul; 160 (1): 186-91 J Biol Chem 1997 Jun 6; 272 (23): 14532-41
4. Noah TL, Black HR, Cheng PW, et al. Nasal and bronchoalveolar lavage fluid 28. Klatt P, Lamas S. Regulation of protein function by S-glutathiolation in response to
cytokines in early cystic fibrosis. J Infect Dis 1997 Mar; 175 (3): 638-47 oxidative and nitrosative stress. Eur J Biochem 2000 Aug; 267 (16): 4928-44
5. Osika E, Cavaillon JM, Chadelat K, et al. Distinct sputum cytokine profiles in 29. Thomas JA, Mallis RJ. Aging and oxidation of reactive protein sulfhydryls. Exp
cystic fibrosis and other chronic inflammatory airway diseases. Eur Respir J Gerontol 2001 Sep; 36 (9): 1519-26
1999 Aug; 14 (2): 339-46 30. Buhl R, Meyer A, Vogelmeier C. Oxidant-protease interaction in the lung: pros-
6. Tabary O, Escotte S, Couetil JP, et al. Genistein inhibits constitutive and inducible pects for antioxidant therapy. Chest 1996 Dec; 110 (6 Suppl.): 267S-72S
NFkappaB activation and decreases IL-8 production by human cystic fibrosis 31. Gillissen A, Birrer P, McElvaney NG, et al. Recombinant secretory leukoprotease
bronchial gland cells. Am J Pathol 1999 Aug; 155 (2): 473-81 inhibitor augments glutathione levels in lung epithelial lining fluid. J Appl
7. Dai Y, Dean TP, Church MK, et al. Desensitisation of neutrophil responses by Physiol 1993 Aug; 75 (2): 825-32
systemic interleukin 8 in cystic fibrosis. Thorax 1994 Sep; 49 (9): 867-71 32. Vogelmeier C, Biedermann T, Maier K, et al. Comparative loss of activity of
8. Bonfield TL, Konstan MW, Berger M. Altered respiratory epithelial cell cytokine recombinant secretory leukoprotease inhibitor and alpha 1-protease inhibitor
production in cystic fibrosis. J Allergy Clin Immunol 1999; 104 (1): 72-8 caused by different forms of oxidative stress. Eur Respir J 1997 Sep; 10 (9):
9. Pilewski JM, Frizzell RA. Role of CFTR in airway disease. Physiol Rev 1999 Jan; 2114-9
79 Suppl. 1: S215-55 33. Barbero GJ. Therapeutic approaches to cystic fibrosis. Bull World Health Organ
10. van der Vliet A, Eiserich JP, Marelich GP, et al. Oxidative stress in cystic fibrosis: 1994; 72 (3): 341-52
does it occur and does it matter? Adv Pharmacol 1997; 38: 491-513 34. Brenneisen P, Briviba K, Wlaschek M, et al. Hydrogen peroxide (H2O2) increases
11. Tamada T, Hug M, Peters K, et al. Bicarbonate secretion in airway serous cells. the steady-state mRNA levels of collagenase/MMP-1 in human dermal fibro-
Pediatr Pulmonol 2001 Oct; 22: 119-20 blasts. Free Radic Biol Med 1997; 22 (3): 515-24
12. Coakley RD. Regulation of airway surface liquid pH in cystic fibrosis. Pediatr
35. Ogino T, Packer L, Maguire JJ. Neutrophil antioxidant capacity during the respira-
Pulmonol 2001 Oct; 22: 120-1
tory burst: loss of glutathione induced by chloramines. Free Radic Biol Med
13. McShane D, Davies JC, Davies MG, et al. Airway surface pH in subjects with 1997; 23 (3): 445-52
cystic fibrosis. Eur Respir J 2003; 21 (1): 37-42
14. Jayaraman S, Joo NS, Reitz B, et al. Submucosal gland secretions in airways from 36. Bilzer M, Lauterberg BH. Glutathione metabolism in activated human neutrophils:
cystic fibrosis patients have normal [NA(+)] and pH but elevated viscosity. stimulation of glutathione synthesis and consumption of glutathione by reactive
Proc Natl Acad Sci U S A 2001; 98 (14): 8119-23 oxygen species. Eur J Clin Invest 1991 Jun; 21 (3): 316-22
2004 Adis Data Information BV. All rights reserved. Treat Respir Med 2004; 3 (6)
This material is
the copyright of the
original publisher.
Unauthorised copying
and distribution
is prohibited.
360 Hudson
37. Vanglarik CJ, Giron-Calle J, Matalon S, et al. Extracellular glutathione protects 60. Houtmeyers E, Gosselink R, Gayan-Ramirez G, et al. Regulation of mucociliary
airway epithelial cells against oxidative damage caused by hypochlorous acid clearance in health and disease. Eur Respir J 1999 May; 13 (5): 1177-88
exposure. Pediatr Pulmonol 2001 Oct; 22: 276-7 61. King M, Rubin BK. Mucus-controlling agents: past and present. Respir Care Clin
N Am 1999 Dec; 5 (4): 575-94
38. Freeman ML, Huntley SA, Meredith MJ, et al. Destabilization and denaturation of
cellular protein by glutathione depletion. Cell Stress Chaperones 1997 Sep; 2 62. Houtmeyers E, Gosselink R, Gayan-Ramirez G, et al. Effects of drugs on mucus
(3): 191-8 clearance. Eur Respir J 1999 Aug; 14 (2): 452-67
63. Connolly MA. Mucolytics and the critically ill patient: help or hindrance? AACN
39. Calabreses V, Testa G, Ravagna A, et al. HSP70 induction in the brain following
Clin Issues 1995 May; 6 (2): 307-15
ethanol administration in the rat: regulation by glutathione redox state. Biochem
Biophys Res Commun 2000 Mar 16; 269 (2): 397-400 64. Clarke SW. Rationale of airway clearance. Eur Respir J Suppl 1989 Jul; 7: 599s-
603s
40. Liu H, Lightfoot R, Stevens JL. Activation of heat shock factor by alkylating
agents is triggered by glutathione depletion and oxidation of protein thiols. J 65. Jiang N, Dreher KL, Dye JA, et al. Residual oil fly ash induces cytotoxicity and
Biol Chem 1996 Mar 1; 271 (9): 4805-12 mucin secretion by guinea pig tracheal epithelial cells via an oxidant-mediated
mechanism. Toxicol Appl Pharmacol 2000 Mar 15; 163 (3): 221-30
41. Wilhelm D, Bender K, Knebel A, et al. The level of intracellular glutathione is a
66. Li JD, Feng W, Gallup M, et al. Activation of NF-kappaB via a Src-dependent Ras-
key regulator for the induction of stress-activated signal transduction pathways
MAPK-pp90rsk pathway is required for Pseudomonas aeruginosa-induced
including Jun N-terminal protein kinases and p38 kinase by alkylating agents.
mucin overproduction in epithelial cells. Proc Natl Acad Sci U S A 1998; 95
Mol Cell Biol 1997 Aug; 17 (8): 4792-800
(10): 5718-23
42. Kirlin WG, Cai J, Thompson SA, et al. Glutathione redox potential in response to
67. Fischer B, Voynow J. Neutrophil elastase induces MUC5AC messenger RNA
differentiation and enzyme inducers. Free Radic Biol Med 1999 Dec; 27 (11-
expression by an oxidant-dependent mechanism. Chest 2000 May; 117 (5
12): 1208-18
Suppl. 1): 317S-20S
43. Lertratanangkoon K, Savaraj N, Scimeca JM, et al. Glutathione depletion-induced 68. Day BJ, van Heeckeren A, Velsor LW. Elevation of lung CFTR, MRP-2, and
thymidylate insufficiency for DNA repair synthesis. Biochem Biophys Res epithelial lining fluid glutathione in mice with Pseudomonas aeruginosa lung
Commun 1997 May 19; 234 (2): 470-5 infection [abstract]. Am J Respir Crit Care Med 2003 Apr; 167 (7): A916
44. Asensi M, Garcia-Espana A, Pallardo FV, et al. Effect of nonprotein thiols on 69. Day BJ, van Heeckeren AM, Min E, et al. Role for cystic fibrosis transmembrane
protein synthesis in isolated rat hepatocytes. Experientia 1996 Feb 15; 52 (2): conductance regulator protein in a glutathione response to bronchopulmonary
111-4 pseudomonas infection. Infect Immun 2004 Apr; 72 (4): 2045-51
45. Fussell JC, Kelly FJ. Effects of dexamethasone on lung protein turnover. Biochem 70. Boxer LA, Oliver JM, Speilberg SP, et al. Protection of granulocytes by vitamin E
J 1991 Jan 1; 273 (Pt 1): 93-7 in glutathione synthetase deficiency. N Engl J Med 1979 Oct 25; 301 (17): 901-
46. Murthy MR. Protein synthesis in growing-rat tissues: II. Polyribosome concentra- 5
tion of brain and liver as a function of age. Biochim Biophys Acta 1966 Jun 22; 71. Cho S, Urata Y, Ilada T, et al. Glutathione downregulates the phosphorylation of I
119 (3): 599-613 kappa-B: autoloop regulation of the NF-kappa B-mediated expression of NF-
47. Thanislass J, Raveendran M, Sivasithamparam N, et al. Effect of chronic gluta- kappa B subunits by TNF-alpha in mouse vascular endothelial cells. Biochem
thione deficiency on rat lung mitochondrial function. Pulm Pharmacol 1996 Biophys Res Commun 1998; 253 (1): 104-8
Apr; 9 (2): 119-22 72. Droge W, Eck HP, Gmunder H, et al. Modulation of lymphocyte functions and
48. Rojas E, Valverde M, Kala SV, et al. Accumulation of DNA damage in the organs immune responses by cysteine and cysteine derivatives. Am J Med 1991 Sep
of mice deficient in gamma-glutamyltranspeptidase. Mutat Res 2000 Feb 14; 30; 91 (3C): 140-3
447 (2): 305-16 73. Sen CK, Khanna S, Reznick AZ, et al. Glutathione regulation of tumor necrosis
49. Gilmont RR, Dardano A, Young M, et al. Effects of glutathione depletion on factor-alpha-induced NF-kappa B activation in skeletal muscle-derived L6
oxidant-induced endothelial cell injury. J Surg Res 1998 Nov; 80 (1): 62-8 cells. Biochem Biophys Res Commun 1997 Aug 28; 237 (3): 645-9
50. Fiorentini C, Falzano L, Rivabene R, et al. N-acetylcysteine protects epithelial cells 74. Desai A, Huang X, Warren JS. Intracellular glutathione redox status modulates
against the antioxidant imbalance due to Clostridium difficile toxins. FEBS Lett MCP-1 expression in pulmonary granulomatous vasculitis. Lab Invest 1999 Jul;
1999 Jun 18; 453 (1-2): 124-8 79 (7): 837-47
51. Kamei H. Cystine starvation induces reversible large-body formation from nuclear 75. Luo Y, Hattori A, Munoz J, et al. Intrastriatal dopamine injection induces apoptosis
bodies in T24 cells. Exp Cell Res 1997 Nov 25; 237 (1): 207-16 through oxidation-involved activation of transcription factors AP-1 and NF-
52. Zuelke KA, Jones DP, Perreault SD. Glutathione oxidation is associated with kappaB in rats. Mol Pharmacol 1999 Aug; 56 (2): 254-64
altered microtubule function and disrupted fertilization in mature hamster 76. Haddad JJ, Olver RE, Land SC. Antioxidant/pro-oxidant equilibrium regulates
oocytes. Biol Reprod 1997 Dec; 57 (6): 1413-9 HIF-1alpha and NF-kappaB redox sensitivity: evidence for inhibition by gluta-
53. Jain A, Martensson J, Mehta T, et al. Ascorbic acid prevents oxidative stress in thione oxidation in alveolar epithelial cells. J Biol Chem 2000; 275 (28): 21130-
glutathione deficient mice: effects on lung type 2 cell lamellar bodies, lung 9
surfactant, and skeletal muscle. Proc Natl Acad Sci U S A 1992 Jun; 89 (11): 77. Tanaka C, Kamata H, Takeshita H, et al. Redox regulation of lipopolysaccharide
5093-7 (LPS)-induced interleukin-8 (IL-8) gene expression mediated by NF kappa B
54. Shaheen SO, Sterne JA, Songhurst CE, et al. Frequent paracetamol use and asthma and AP-1 in human astrocytoma U373 cells. Biochem Biophys Res Commun
in adults. Thorax 2000 Apr; 55 (4): 266-70 1997 Mar 17; 232 (2): 568-73
55. Li XY, Donaldson K, Rahman I, et al. An investigation of the role of glutathione in 78. Rahman I, MacNee W. Regulation of redox glutathione levels and gene transcrip-
increased epithelial permeability induced by cigarette smoke in vivo and in tion in lung inflammation: therapeutic approaches. Free Radic Biol Med 2000
vitro. Am J Respir Crit Care Med 1994 Jun; 149 (6): 1518-25 May 1; 28 (9): 1405-20
56. Remiao F, Carmo H, Carvalho FD, et al. Inhibition of glutathione reductase by 79. Christman JW, Sadikot RT, Blackwell TS. The role of nuclear factor-kappa B in
isoproterenol oxidation products. J Enzyme Inhib 2000; 15 (1): 47-61 pulmonary diseases. Chest 2000; 117 (5): 1482-7
57. Barker JE, Heales SJ, Cassidy A, et al. Depletion of brain glutathione results in a 80. Gosset P, Wallaert B, Tonnel AB, et al. Thiol regulation of the production of TNF-
decrease of glutathione reductase activity: an enzyme susceptible to oxidative alpha, IL-6 and IL-8 by human alveolar macrophages. Eur Respir J 1999 Jul; 14
damage. Brain Res 1996 Apr 15; 16 (1-2): 118-22 (1): 98-105
58. van Klaveren RJ, Hoet PH, Pype JL, et al. Increase in gamma-glutamyltransferase 81. Hashimoto S, Gon Y, Matsumoto K, et al. Regulation by intracellular glutathione
by glutathione depletion in rat type II pneumocytes. Free Radic Biol Med 1997; of TNF-alpha-induced p38 MAP kinase activation and RANTES production by
22 (3): 525-34 human pulmonary vascular endothelial cells. Allergy 2000 May; 55 (5): 463-9
59. Smith CV, Jones DP, Guenthner TM, et al. Compartmentation of glutathione: 82. Yamauchi N, Watanabe N, Kuriyama H, et al. Suppressive effects of intracellular
implications for the study of toxicity and disease. Toxicol Appl Pharmacol glutathione on hydroxyl radical production induced by tumor necrosis factor.
1996; 140: 1-12 Int J Cancer 1990 Nov 15; 46 (5): 884-8
2004 Adis Data Information BV. All rights reserved. Treat Respir Med 2004; 3 (6)
This material is
the copyright of the
original publisher.
Unauthorised copying
and distribution
is prohibited.
Pathogenesis of Cystic Fibrosis 361
83. Peristeris P, Clark BD, Gatti S, et al. N-acetylcysteine and glutathione as inhibitors 107. Smyth MJ. Glutathione modulates activation-dependent proliferation of human
of tumor necrosis factor production. Cell Immunol 1992 Apr; 140 (2): 390-9 peripheral blood lymphocyte populations without regulating their activated
function. J Immunol 1991 Mar 15; 146 (6): 1921-7
84. Rovin BH, Dickerson JA, Tan LC, et al. Modulation of IL-1-induced chemokine
expression in human mesangial cells through alterations in redox status. 108. Romero DL, Mounho BJ, Lauer FT, et al. Depletion of glutathione by benzo
Cytokine 1997 Mar; 9 (3): 178-86 (a)pyrene metabolites, ionomycin, thapsigargin, and phorbol myristate in
human peripheral blood mononuclear cells. Toxicol Appl Pharmacol 1997
85. Pena LR, Hill DB, McClain CJ. Treatment with glutathione precursor decreases May; 144 (1): 62-9
cytokine activity. JPEN J Parenter Enteral Nutr 1999 Jan-Feb; 23 (1): 1-6
109. Liang SM, Liang CM, Hargrove ME, et al. Regulation by glutathione of the effect
86. Watson RW, Rotstein OD, Nathens AB, et al. Thiol-mediated redox regulation of
of lymphokines on differentiation of primary activated lymphocytes: influence
neutrophil apoptosis. Surgery 1996 Aug; 120 (2): 150-7
of glutathione on cytotoxic activity of CD3-AK-. J Immunol 1991 Mar 15; 146
87. Nakatani T, Tawaramoto M, Opare Kennedy D, et al. Apoptosis induced by (6): 1909-13
chelation of intracellular zinc is associated with depletion of cellular reduced
110. Gmunder H, Droge W. Differential effects of glutathione depletion on T cell
glutathione level in rat hepatocytes. Chem Biol Interact 2000 Mar 15; 125 (3):
subsets. Cell Immunol 1991 Nov; 138 (1): 229-37
151-63
111. Ting CC, Hargrove HE, Liang SM, et al. Dichotomy of glutathione regulation of
88. Coppola S, Ghibelli L. GSH extrusion and the mitochondrial pathway of apoptotic
the activation of resting and preactivated lymphocytes. Cell Immunol 1992 Jun;
signalling. Biochem Soc Trans 2000 Feb; 28 (2): 56-61
142 (1): 40-53
89. Colell A, Garcia-Ruiz C, Miranda M, et al. Selective glutathione depletion of
112. Gmunder H, Eck HP, Benninghoff B, et al. Macrophages regulate intracellular
mitochondria by ethanol sensitizes hepatocytes to tumor necrosis factor. Gas-
glutathione levels of lymphocytes: evidence for an immunoregulatory role of
troenterology 1998 Dec; 115 (6): 1541-51
cysteine. Cell Immunol 1990 Aug; 129 (1): 32-46
90. Nicole A, Santiard-Baron D, Ceballos-Picot I. Direct evidence for glutathione as
113. Galietta LJV, Folli C, Marchetti C, et al. Modification of transepithelial ion
mediator of apoptosis in neuronal cells. Biomed Pharmacother 1998; 52 (9):
transport in human cultured bronchial epithelial cells by interferon-gamma. Am
349-55
J Physiol Lung Cell Mol Physiol 2000 Jun; 278 (6): L1186-94
91. Rosenfeld ME. Inflammation, lipids, and free radicals: lessons learned from the
114. van der Meide PH, de Labie MC, Botman CA, et al. Mercuric chloride down-
atherogenic process. Semin Reprod Endrocrinol 1998; 16 (4): 249-61
regulates T cell interferon-gamma production in brown Norway but not in
92. Hardwick SJ, Carpenter KL, Allen EA, et al. Glutathione (GSH) and the toxicity of Lewis rats: role of glutathione. Eur J Immunol 1993 Mar; 23 (3): 675-81
oxidised low-density lipoprotein to human monocyte-macrophages. Free Radic
115. Sprietsma JE. Zinc-controlled Th1/Th2 switch significantly determines develop-
Res 1999 Jan; 30 (1): 11-9
ment of disease. Med Hypotheses 1997 Jul; 49 (1): 1-14
93. Aoshiba K, Yasui S, Nishimura K, et al. Thiol depletion induces apoptosis in
116. Sprietsma JE. Cysteine, glutathione (GSH) and zinc and copper ions together are
cultured lung fibroblasts. Am J Respir Cell Mol Biol 1999 Jul; 21 (1): 54-64
effective, natural, intracellular inhibitors of (AIDS) viruses. Med Hypotheses
94. Vahrmeijer AL, van Dierendonck JH, Schutrups J, et al. Effect of glutathione 1999 Jun; 52 (6): 529-38
depletion on inhibition of cell cycle progression and induction of apoptosis by
117. Spriestma JE. Modern diets and diseases: NO-zinc balance: under Th1, zinc and
melphalan (L-phenylalanine mustard) in human colorectal cancer cells. Bi-
nitrogen monoxide (NO) collectively protect against viruses, AIDS, autoim-
ochem Pharmacol 1999 Aug 15; 58 (4): 655-64
munity, diabetes, allergies, asthma, infectious diseases, atherosclerosis and
95. Yang CF, Shen HM, Ong CN. Ebselen induces apoptosis in HepG (2) cells through cancer. Med Hypotheses 1999 Jul; 53 (1): 6-16
rapid depletion of intracellular thiols. Arch Biochem Biophys 2000 Feb 15; 374
(2): 142-52 118. Moser C, Johansen HK, Song Z, et al. Chronic pseudomonas aeruginosa lung
infection is more severe in Th2 responding BALB/c mice compared to Th1
96. Li Y, Maher P, Schubert D. A role for 12-lipoxygenase in nerve cell death caused responding C3H/HeN mice. APMIS 1997 Nov; 105 (11): 838-42
by glutathione depletion. Neuron 1997 Aug; 19 (2): 453-63
119. Chen G, Wang SH, Warner TD. Regulation of iNOS mRNA levels in endothelial
97. Iwata S, Hori T, Sato N, et al. Adult T cell leukemia (ATL)-derived factor/human
cells by glutathione, a double-edged sword. Free Radic Res 2000 Mar; 32 (3):
thioredoxin prevents apoptosis of lymphoid cells induced by L-cystine and
223-34
glutathione depletion: possible involvement of thiol-mediated redox regulation
in apoptosis caused by pro-oxidant state. J Immunol 1997 Apr; 158 (7): 3108-17 120. Kang KW, Pak YM, Kim ND. Diethylmaleate and buthionine sulfoximine, gluta-
thione-depleting agents, differentially inhibit expression of inducible nitric
98. Wedner HJ, Simchowitz L, Stenson WF, et al. Inhibition of human
oxide synthase in endotoxemic mice. Nitric Oxide 1999 Jun; 3 (3): 265-71
polymorphonuclear leukocyte function by 2-cyclohexene-1-one: A role for
glutathione in cell activation. J Clin Invest 1981 Aug; 68 (2): 535-43 121. Vos TA, Goor H, Tuyt L, et al. Expression of inducible nitric oxide synthase in
endotoxemic rat hepatocytes is dependent on the cellular glutathione status.
99. Aziz AS, Klesius PH, Frandsen JC. Effects of selenium on polymorphonuclear
Hepatology 1999 Feb; 29 (2): 421-6
leukocyte function in goats. Am J Vet Res 1984 Sep; 45 (9): 1715-8
122. Wang F, Wang LY, Wright D, et al. Redox imbalance differentially inhibits
100. Voetman AA, Loos JA, Roos D. Changes in the levels of glutathione in phago-
lipopolysaccharide-induced macrophage activation in the mouse liver. Infect
cytosing human neutrophils. Blood 1980 May; 55 (5): 741-7
Immun 1999 Oct; 67 (10): 5409-16
101. Peterson JD, Herzenberg LA, Vasquez K, et al. Glutathione levels in antigen-
123. Davidson CA, Kaminski PM, Wolin MS. NO elicits prolonged relaxation of bovine
presenting cells modulate Th1 versus Th2 response patterns. Proc Natl Acad Sci
pulmonary arteries via endogenous peroxynitrite generation. Am J Physiol 1997
U S A 1998 Mar 17; 95 (6): 3071-6
Aug; 273 (2 Pt 1): L437-44
102. Nathens AB, Rotstein OD, Dackiw AP, et al. The glutathione depleting agent
124. Singh SP, Wishnok JS, Keshive M, et al. The chemistry of the S-nitrosogluta-
diethylmaleate prolong renal allograft survival. J Surg Res 1998 Jun; 77 (1): 75-
thione/glutathione system. Proc Natl Acad Sci U S A 1996 Dec; 93: 14428-33
9
125. D’Emilia DM, Lipton SA. Ratio of S-nitrosohomocyst(e)ine or other thiols deter-
103. Robinson MK, Rodrick ML, Jacobs DO, et al. Glutathione depletion in rats impairs
mines neurotoxicity in rat cerebrocortical cultures. Neurosci Lett 1999; 265:
T-cell and macrophage immune function. Arch Surg 1993 Jan; 128 (1): 29-34
103-6
104. Ginn-Pease ME, Whisler RL. Optimal NF kappa B mediated transcriptional
126. Stefanelli C, Pignatti C, Tantani B, et al. Nitric oxide can function as either a killer
responses to Jurkat T cells exposed to oxidative stress are dependent on
molecule or an antiapoptotic effector in cardiomyocytes. Biochim Biophys Acta
intracellular glutathione and costimulatory signals. Biochem Biophys Res
1999 Jul 8; 1450 (3): 406-13
Commun 1996 Sep 24; 226 (3): 695-702
127. Rosenberg PA, Li Y, Ai S, et al. Intracellular redox state determines whether nitric
105. Jeannin P, Delneste Y, Lecoanet-Henchoz S, et al. Thiols decrease human in-
oxide is toxic or protective to rat oligodendrocytes in culture. J Neurochem
terleukin (IL) 4 production and IL-4-induced immunoglobulin synthesis. J Exp
1999 Aug; 73 (2): 476-84
Med 1995 Dec 1; 182 (6): 1785-92
106. Rigacci S, Iantomasi T, Marraccini P, et al. Evidence for glutathione involvement 128. Chiueh CC, Rauhala P. The redox pathway of S-nitrisoglutathione, glutathione and
in platelet-derived growth-factor-mediated signal. Biochem J 1997 Jun 15; 324 nitric oxide in cell to neuron communications. Free Radic Res 1999 Dec; 31 (6):
(Pt 3): 791-6 641-50
2004 Adis Data Information BV. All rights reserved. Treat Respir Med 2004; 3 (6)
This material is
the copyright of the
original publisher.
Unauthorised copying
and distribution
is prohibited.
362 Hudson
129. Jones KL, Bryan TW, Jinkins PA, et al. Superoxide released from neutrophils 155. Tirouvanziam R, Khazaal I, Peault B. Primary inflammation in human cystic
causes a reduction in nitric oxide gas. Am J Physiol 1998 Dec; 275 (6): L1120-6 fibrosis small airways. Am J Physiol Lung Cell Mol Physiol 2002 Aug; 283 (2):
L445-51
130. Zavodnik IB, Lapshina EA, Rekawiecka K, et al. Membrane effects of nitrite-
156. Tirouvanziam R, de Bentzmann S, Hubeau C, et al. Inflammation and infection in
induced oxidation of human red blood cells. Biochim Biophys Acta 1999 Oct
naive human cystic fibrosis airway grafts. Am J Respir Cell Mol Biol 2000
15; 1421 (2): 306-16
Aug; 23 (2): 121-7
131. Kelley TJ, Drumm ML. Inducible nitric oxide synthase expression is reduced in 157. Escotte S, Laplace V, Benoit S, et al. Basal KC production in tracheal surface
cystic fibrosis murine and human airway epithelial cell lines. J Clin Invest 1998 liquid of CF mice engrafted in nude mice [abstract]. Pediatr Pulmonol 2001
Sep; 102 (6): 1200-7 Oct; 22: 227
132. Mayer B, Pfeiffer S, Schrammel A, et al. A new pathway of nitric oxide cyclic 158. Lancellotti L, D’Orazio C, Mastella G, et al. Deficiency of vitamins E and A in
GMP signalling involving s-nitrosoglutathione. J Biol Chem 1998 Feb 6; 273 cystic fibrosis is independent of pancreatic function and current enzyme and
(6): 3264-70 vitamin supplementation. Eur J Pediatr 1996 Apr; 155 (4): 281-5
133. Belvisi MG, Ward JK, Mitchell JA, et al. Nitric oxide as a neurotransmitter in 159. Portal BC, Richard MJ, Faure HS, et al. Altered antioxidant status and increased
human airways. Arch Int Pharmacodyn Ther 1995 Jan-Feb; 329 (1): 97-110 lipid peroxidation in children with cystic fibrosis. Am J Clin Nutr 1995 Apr; 61
134. Gaston B, Drazen JM, Jansen A, et al. Relaxation of human bronchial smooth (4): 843-7
muscle by S-nitrosothiols in vitro. J Pharmacol Exp Ther 1994 Feb; 268 (2): 160. Winklhofer-Roob BM, Ellemunter H, Fruhwirth M, et al. Plasma vitamin C
978-84 concentrations in patients with cystic fibrosis: evidence of associations with
135. Kamosinska B, Radomski MW, Duszyk M, et al. Nitric oxide activates chloride lung inflammation. Am J Clin Nutr 1997 Jun; 65 (6): 1858-66
currents in human lung epithelial cells. Am J Physiol 1997 Jun; 272 (6 Pt 1): 161. Wood LG, Fitzgerald DA, Gibson PG, et al. Oxidative stress in cystic fibrosis:
L1098-104 dietary and metabolic factors. Am Coll Nutr 2001 Apr; 20 (2 Suppl.): 157-65
136. Dong YJ, Chao AC, Kouyama K, et al. Activation of CFTR chloride current by 162. Madarasi A, Lugassi A, Greiner E, et al. Antioxidant status in patients with cystic
nitric oxide in human T lymphocytes. EMBO J 1995 Jun 15; 14 (12): 2700-7 fibrosis. Ann Nutr Metab 2000; 44 (5-6): 207-11
137. Runer T, Lindberg S. Ciliostimulatory effects mediated by nitric oxide. Acta 163. Isabelle D, Puget M, Bellon G, et al. Antioxidant and peroxidative status in 312
Otolaryngol 1999; 119 (7): 821-5 patients with cystic fibrosis. Pediatr Pulmonol 2002 Oct; 24: 339
138. Linsdell P, Hanrahan JW. Glutathione permeability of CFTR. Am J Physiol 1998 164. Birrer P, McElvaney NG, Rudeberg A, et al. Protease-antiprotease imbalance in the
Jul; 44 (1): C323-6 lungs of children with cystic fibrosis. Am J Respir Crit Care Med 1994 Jul; 150
139. Gao L, Kim KJ, Yankaskas JR, et al. Abnormal glutathione transport in cystic (1): 207-13
fibrosis airway epithelia. Am J Physiol 1999 Jul; 277 (1): L113-8 165. Griese M, Birrer P, Demirsoy A. Pulmonary surfactant in cystic fibrosis. Eur
140. Velsor LW, van Heeckeren A, Day BJ. Antioxidant imbalance in the lungs of Respir J 1997 Sep; 10 (9): 1983-8
cystic fibrosis transmembrane conductance regulator protein mutant mice. Am J 166. DiMango E, Tabibi S, Ratner A, et al. Activation of NF-kappaB by adherent
Physiol Lung Cell Mol Physiol 2001 Jul; 281 (1): L31-8 Pseudomonas aeruginosa in normal cystic fibrosis respiratory epithelial cells. J
Clin Investig 1998; 101 (11): 2598-605
141. Kogan I, Ramjeesingh M, Kidd J, et al. Characterization of glutathione permeabili-
ty through the CFTR channel pore. Pediatr Pulmonol 2002 Oct; 24: 189-90 167. Weber A, Nguyen B, Bryan R, et al. Effects of CFTR mutations on epithelial
cytokine expression [abstract]. Pediatr Pulmonol 1999 Sep; Suppl. 19: 309-10
142. Kogan I, Ramjessingh M, Li C, et al. CFTR directly mediates nucleotide-regulated
168. Elborn JS, Cordon SM, Western PJ, et al. Tumor necrosis-factor-alpha, resting
glutathione flux. EMBO J 2003 May 1; 22 (9): 1981-9
energy expenditure and cachexia in cystic fibrosis. Clin Sci (Colch) 1993 Nov;
143. Finder JD, Engman CL, Kagan VE, et al. Bronchoalveolar lavage fluid from 85 (5): 563-8
patients with cystic fibrosis have diminished levels of reduced glutathione 169. Pfeffer KD, Huecksteadt TP, Hoidal JR. Expression and regulation of tumor
[abstract]. Ped Pulm 2003; 239 Suppl. 25: A158 necrosis factor in macrophages from cystic fibrosis patients. Am J Respir Cell
144. Hull J, Vervaart P, Grimwood K, et al. Pulmonary oxidative stress response in Mol Biol 1993 Nov; 9 (5): 511-9
young children with cystic fibrosis. Thorax 1997; 52: 557-60 170. Greally P, Hussain MJ, Vergani D, et al. Serum interleukin-1 alpha and soluble
145. Tirouvanziam R. CFTR, glutathione, and neutrophil function [online]. Available interleukin-2 receptor concentrations in cystic fibrosis. Arch Dis Child 1993
from URL: http://63.193.197.190/Subsets/tirouv/index.htm [Accessed 1999 Jun; 68 (6): 785-7
Nov 13] 171. Wilmott RW, Frenzke M, Kociela V, et al. Plasma interleukin-1 alpha and beta,
146. Neefjes VM, Evelo CT, Baars LG, et al. Erythrocyte glutathione S transferase as a tumor necrosis factor-alpha, and lipopolysaccharide concentrations during pul-
marker of oxidative stress at birth. Arch Dis Child Fetal Neonatal Ed 1999 Sep; monary exacerbations of cystic fibrosis. Pediatr Pulmonol 1994 Jul; 18 (1): 21-
81 (2): F130-3 7
147. Brown RK, Wyatt H, Price JF, et al. Pulmonary dysfunction in cystic fibrosis is 172. Lundberg JO, Nordvall SL, Weitzberg E, et al. Exhaled nitric oxide in paediatric
associated with oxidative stress. Eur Respir J 1996; 9: 334-9 asthma and cystic fibrosis. Arch Dis Child 1996; 75: 323-6
148. Roum JH, Buhl R, McElvaney NG, et al. Systemic deficiency of glutathione in 173. Kroesbergen A, Jobsis Q, Bel EH, et al. Flow-dependency of exhaled nitric oxide
cystic fibrosis. J Appl Physiol 1993 Dec; 75 (6): 2419-24 in children with asthma and cystic fibrosis. Eur Respir J 1999 Oct; 14 (4): 871-5
149. Tirouvanziam RM, Tjioe I, Moss RB, et al. Multiparameter study of CF blood and 174. Grasemann H, Michler E, Wallot M, et al. Decreased concentration of exhaled
lung leukocytes using 3-laser, 11-color cytometry. Pediatr Pulmonol 2001 Oct; nitric oxide (NO) in patients with cystic fibrosis. Pediatr Pulmonol 1997; 24:
22: 271-2 173-7
150. Abraham EH, Sterling KM, Kim RJH, et al. Erythrocyte membrane ATP binding 175. Balfour-Lynn IM, Laverty A, Dinwiddie R. Reduced upper airway nitric oxide in
cassette (ABC) proteins: MRP1 and CFTR as well as CD39 (ectoapyrase) cystic fibrosis. Arch Dis Child 1996; 75: 319-22
involved in RBC ATP transport and elevated blood plasma ATP of cystic 176. Dotsch J, Demirakca S, Terbrack HG, et al. Airway nitric oxide in asthmatic
fibrosis. Blood Cells Mol Dis 2001 Jan-Feb; 27 (1): 165-80 children and patients with cystic fibrosis. Eur Respir J 1996; 9: 2537-40
151. Dominguez C, Gartner S, Linan S, et al. Enhanced oxidative damage in cystic 177. Thomas SR, Kharitonov SA, Scott SF, et al. Nasal and exhaled nitric oxide is
fibrosis patients. Biofactors 1998; 8 (1-2): 149-53 reduced in adult patients with cystic fibrosis and does not correlate with cystic
152. Lloyd-Still JD, Ganther HE. Selenium and glutathione peroxidase levels in cystic fibrosis genotype. Chest 2000 Apr; 117 (4): 1085-9
fibrosis. Pediatrics 1980; 65: 1010-2 178. Ho LP, Innes JA, Greening AP. Nitrite levels in breath condensate of patients with
153. Carmagnol F, Sinet PM, Lenoir G, et al. Absence of modifications of the enzyme cystic fibrosis is elevated in contrast to exhaled nitric oxide. Thorax 1998 Aug;
defense system against oxygen toxicity in cystic fibrosis. Pediatr Res 1983; 17: 53 (8): 680-4
181-2 179. Andersson C, Gaston B, Roomans G. S-Nitrosoglutathione induces functional
154. Konstan MW, Berger M. Current understanding of the inflammatory process in deltaF508-CFTR in airway epithelial cells. Biochem Biophys Res Commun
cystic fibrosis: onset and etiology. Pediatr Pulmnol 1997 Aug; 24 (2): 137-42 2002 Sep 27; 297 (3): 552-7
2004 Adis Data Information BV. All rights reserved. Treat Respir Med 2004; 3 (6)
This material is
the copyright of the
original publisher.
Unauthorised copying
and distribution
is prohibited.
Pathogenesis of Cystic Fibrosis 363
180. Snyder AH, McPherson ME, Hunt JF, et al. Acute effects of aerosolized S- 193. Griese M, Ramakers J, Krasselt A, et al. Improvement of alveolar glutathione and
nitrosoglutathione in cystic fibrosis. Am J Respir Crit Care Med 2002 Apr 1; lung function but not oxidative state in cystic fibrosis. Am J Respir Crit Care
165 (7): 922-6 Med 2004 Apr 1; 169 (7): 822-8
181. Jungas T, Motta I, Duffieux F, et al. Glutathione levels and BAX activation during 194. Bishop C, Hudson VM, Hilton SC, et al. Effect of inhaled buffered glutathione
apoptosis due to oxidative stress in cells expressing wild-type and mutant cystic (GSH) on the clinical status of cystic fibrosis patients [abstract]. Am J Respir
fibrosis transmembrane conductance regulator. J Biol Chem 2002 Aug 2; 277 Crit Care Med 2003 Apr; 167 (7): A918
(31): 27912-8 195. Bishop CT, Hudson VM, Hilton SC, Wilde C. A pilot study of the effect of inhaled
182. Hosseini Nia T, Kubow S, Grey V, et al. Inhibition of Pseudomonas aeroginosa-buffered reduced glutathione on the clinical status of cystic fibrosis patients.
mediated mortality and weight loss via ingestion of whey treated by hyperbaric Chest. In press
pressure [abstract]. Pediatr Pulmonol 2002 Oct; 24: 276 196. Aw TY, Wierzbicka G, Jones DP. Oral glutathione increases tissue glutathione in
183. Buhl R, Vogelmeier C, Critenden M, et al. Augmentation of the glutathione in the rats. Chem Biol Interact 1991; 80 (1): 89-97
fluid lining the epithelium of the lower respiratory tract by directly administer- 197. Iantomasi T, Favilli F, Marraccini P, et al. Glutathione transport system in human
ing glutathione. Proc Natl Acad Sci U S A 1990 Jun; 87: 4063-7 small intestine epithelial cells. Biochim Biophys Acta 1997 Dec 4; 1330 (2):
184. Buhl R, Holroyd K, Borok Z, et al. Reversal of the glutathione deficiency in the 274-83
lower respiratory tract of HIV-seropositive individuals by glutathione aerosol 198. Hunjan MK, Evered DF. Absorption of glutathione from the gastro-intestinal tract.
therapy [abstract]. Clin Res 1990; 38 (2): 596A
Biochim Biophys Acta 1985 May 14; 815 (2): 184-8
185. Holroyd KJ, Buhl R, Borok Z, et al. Correction of glutathione deficiency in the 199. Favilli F, Marracini P, Iantomasi T, et al. Effect of orally administered glutathione
lower respiratory tract of HIV seropositive individuals by glutathione aerosol
on glutathione levels in some organs of rats: role of specific transporters. Br J
treatment. Thorax 1993 Oct; 48 (10): 985-9
Nutr 1997 Aug; 78 (2): 293-300
186. Borok Z, Buhl R, Grimes GJ, et al. Effect of glutathione aerosol on oxidant- 200. Hagen TM, Wierzbicka GT, Sillau AH, et al. Bioavailability of dietary glutathione:
antioxidant imbalance in idiopathic pulmonary fibrosis. Lancet 1991 Jul 17;
effect on plasma concentration. Am J Physiol 1990 Oct; 259 (4 Pt 1): G524-9
338: 215-6
201. Gao L, Broughman JR, Iwamoto T, et al. Synthetic chloride channel restores
187. Marrades RM, Roca J, Barbera JA, et al. Nebulized glutathione induces
glutathione secretion in cystic fibrosis airway epithelia. Am J Physiol Lung Cell
bronchoconstriction in patients with mild asthma. Am J Respir Crit Care Med
1997; 156: 425-30 Mol Physiol 2001 Jul; 281 (1): L24-30
202. Ciuchi E, Odetti P, Prando R. The effect of acute glutathione treatment on sorbitol
188. Bernorio S, Pecis M, Zucchi A, et al. Glutathione in bronchial hyperresponsive-
ness. J Aerosol Med 1996; 9 (2): 207-13 level in erythrocytes from diabetic patients. Diabetes Metab 1997 Feb; 23 (1):
58-60
189. Testa B, Mesolella M, Testa D, et al. Glutathione in the upper respiratory tract. Ann
203. Sechi G, Deledda MG, Bua G, et al. Reduced intravenous glutathione in the
Otol Rhinol Laryngol 1995; 104: 117-9
treatment of early Parkinson’s disease. Prog Neuropsychopharmacol Biol Psy-
190. Roum JH, Borok Z, McElvaney NG, et al. Glutathione aerosol suppresses lung chiatry 1996 Oct; 20 (7): 1159-70
epithelial surface inflammatory cell-derived oxidants in cystic fibrosis. J Appl
Physiol 1999 Jul; 87 (1): 438-43 204. Visca AG. Adherence to treatment and self-prescription on new drugs: an emerging
problem [abstract]. Pediatr Pulmonol 2002 Oct; 24: 347
191. Bagnato GF, Gulli S, De Pasquale R, et al. Effect of inhaled glutathione on airway
response to ‘Fog’ challenge in asthmatic patients. Respiration 1999 Nov-Dec;
66 (6): 518-21 Correspondence and offprints: Dr Valerie M. Hudson, Brigham Young Uni-
192. Griese M, Ramakers J, Krasselt A, et al. Optimized aerosol delivery of glutathione versity, 760 SWKT, Provo, UT 84602, USA.
(GSH) to patients with cystic fibrosis (CF) increases alveolar GSH concentra-
tion [abstract]. Am J Respir Crit Care Med 2003 Apr; 167 (7): A919 E-mail: valerie_hudson@byu.edu
2004 Adis Data Information BV. All rights reserved. Treat Respir Med 2004; 3 (6)
Article
Full-text available
Bicarbonate is one of the major anions in mammalian tissues and fluids, is utilized by various exchangers to transport other ions and organic substrates across cell membranes and plays a critical role in cell and systemic pH homeostasis. Chloride/bicarbonate (Cl−/HCO3−) exchangers are abundantly expressed in erythrocytes and epithelial cells and, as a consequence, are particularly exposed to oxidants in the systemic circulation and at the interface with the external environment. Here, we review the physiological functions and pathophysiological alterations of Cl−/HCO3− exchangers belonging to the solute carriers SLC4 and SLC26 superfamilies in relation to oxidative stress. Particularly well studied is the impact of oxidative stress on the red blood cell SLC4A1/AE1 (Band 3 protein), of which the function seems to be directly affected by oxidative stress and possibly involves oxidation of the transporter itself or its interacting proteins, with detrimental consequences in oxidative stress‐related diseases including inflammation, metabolic dysfunctions, and aging. The effect of oxidative stress on SLC26 members was less extensively explored. Indirect evidence suggests that SLC26 transporters can be target as well as determinants of oxidative stress, especially when their expression is abolished or dysregulated.
Article
Full-text available
Enterotoxigenic Escherichia coli (ETEC) is a major diarrheal pathogen in children in low- to middle-income countries, deployed military personnel, and travelers to regions of endemicity. The heat-stable toxin (ST) is a small nonimmunogenic secreted peptide with 3 disulfide bonds. It has been appreciated that dietary disulfides modulate intestinal redox potential and that ST could be detoxified using exogenous reductants. Using biochemical and spectroscopic approaches, we demonstrated that ST can separately bind iron and zinc under reducing conditions, thereby reducing ST toxicity. Moreover, we demonstrated that ST modulates the glutathione (GSH)/oxidized glutathione (GSSG) ratio and that ST should be considered a toxin oxidant. ST can be detoxified by oxidizing zinc-loaded metallothionine, causing free zinc to be released. These studies help lay a foundation to understand how diarrheal pathogens modulate intestinal redox potential and may impact how we design therapeutics and/or vaccines for the pathogens that produce them.
Article
Full-text available
Cystic fibrosis (CF) occurs when the cystic fibrosis transmembrane conductance regulator (CFTR) protein is not synthetized and folded correctly. The CFTR protein helps to maintain the balance of salt and water on many body surfaces, such as the lung surface. When the protein is not working correctly, chloride becomes trapped in cells, then water cannot hydrate the cellular surface and the mucus covering the cells becomes thick and sticky. Furthermore, a defective CFTR appears to produce a redox imbalance in epithelial cells and extracellular fluids and to cause an abnormal generation of reactive oxygen species: as a consequence, oxidative stress has been implicated as a causative factor in the aetiology of the process. Moreover, massive evidences show that defective CFTR gives rise to extracellular GSH level decrease and elevated glucose concentrations in airway surface liquid (ASL), thus encouraging lung infection by pathogens in the CF advancement. Recent research in progress aims to rediscover a possible role of mitochondria in CF. Here the latest new and recent studies on mitochondrial bioenergetics are collected. Surprisingly, they have enabled us to ascertain that mitochondria have a leading role in opposing the high ASL glucose level as well as oxidative stress in CF.
Article
Full-text available
Glutathione (γ-l-glutamyl-l-cysteinylglycine) is a small tripeptide found at millimolar concentrations in nearly all eukaryotes as well as many prokaryotic cells. Glutathione synthesis is restricted to the cytosol in animals and fungi and to the cytosol and plastids in plants. Nonetheless, glutathione is found in virtually all subcellular compartments. This implies that transporters must exist that facilitate glutathione transport into and out of the various subcellular compartments. Glutathione may also be exported and imported across the plasma membrane in many cells. However, in most cases, the molecular identity of these transporters remains unclear. Whilst glutathione transport is essential for the supply and replenishment of subcellular glutathione pools, recent evidence supports a more active role for glutathione transport in the regulation of subcellular glutathione redox homeostasis. However, our knowledge of glutathione redox homeostasis at the level of specific subcellular compartments remains remarkably limited and the role of glutathione transport remains largely unclear. In this review, we discuss how new tools and techniques have begun to yield insights into subcellular glutathione distribution and glutathione redox homeostasis. In particular, we discuss the known and putative glutathione transporters and examine their contribution to the regulation of subcellular glutathione redox homeostasis.
Article
Progressive lung function decline and ultimately respiratory failure is the most common cause of death in cystic fibrosis (CF). This decline is punctuated by acute pulmonary exacerbations (APEs) and in many cases there is failure to return to baseline lung function. Ultraperformance liquid chromatography-quadrupole-time-of-flight mass spectrometry was used to profile metabolites in exhaled breath condensate (EBC) samples from 17 clinically stable CF patients, 9 CF patients with an APE severe enough to require hospitalization (termed APE), 5 CF patients during recovery from a severe APE (termed post-APE), and 4 CF patients who were clinically stable at the time of collection but in the subsequent 1 to 3 months developed a severe APE (termed pre-APE). A panel containing 2 metabolic discriminant features identified as 4-hydroxycyclohexylcarboxylic acid and pyroglutamic acid differentiated the APE from the stable CF samples with 84.6% accuracy. Pre-APE samples were distinguished from stable CF samples by lactic acid and pyroglutamic acid with 90.5% accuracy, and in general matched the “APE signature” when projected into the APE vs. stable CF model. Post-APE samples were on average more similar to stable CF samples in terms of their metabolomic signature. These results show feasibility for detecting and predicting an oncoming APE, or monitoring APE treatment using EBC metabolites.
Article
Formation of reactive oxygen species (ROS) is a natural process during oxidative metabolism. ROS play an important role not only in pathological processes of human organism as usually presented but less attention is paid to their proper important role in cell signaling, biosynthesis or non-specific antiinfectious defence. Overproduction of ROS during numerous pathological situations in presence of insufficient antioxidant protection leads to substantial oxidative changes of lipids, proteins, sugars, and also DNA. Protection against ROS is assured by different extracellular or intracellular antioxidant mechanisms as studied during last decades. Antioxidant enzymes rectifying the oxidative damage are studied with regard to their different activities and usefulness in body protection. Their genetic polymorphisms are certainly involved in different response to oxidative stress. Special attention should be devoted to the topic of oxidative nuclear and mitochondrial DNA damage and its restoring via DNA repair process, especially base excision repair (BER). A large scale of antioxidant enzymes is involved in correction of DNA oxidative damage. Natural trend of worsened DNA repair is usually associated with aging. Other pathologies related with deficient DNA repair are susceptibility to carcinogenesis (lack of apoptosis control) or degenerative diseases. Oxidative stress is involved in the pathophysiology of diabetes mellitus (DM-oxidative stress of mainly metabolic origin) and inflammatory bowel diseases (IBD-oxidative stress of mainly inflammatory origin). In spite of confirmed OS in DM or IBD, the substantial information about the intensity of DNA repair and its possible relationship to the disease course and development of chronic complications is missing. Our pilot studies completed both in adult and pediatric patients with DM or IBD confirmed an increased oxidative stress as well as oxidative DNA damage examined with comet assay. The surprising findings were ascertained in intensity of DNA repair (analysed with modified comet assay). DNA repair process was stimulated in Type 1 diabetes adults without diabetic microvascular complications and still more in diabetic children with short disease duration. On the other hand adults with Type 2 diabetes had substantially increased oxidative DNA damage and extremely low DNA repair. This finding could be the link to increased susceptibility of Type 2 DM patients to cancerogenesis. Patients with IBD (Crohns̀ disease-both children and adults) had similar tendencies in OS intensity and oxidative DNA damage and repair but less intensive. Large population studies in DM or IBD studying intensity of OS and expression of DNA repair enzymes are needed in order to get the correlation between individual repair enzymes expression and the long-term course and occurence of complications in DM or IBD.
Article
Cystic Fibrosis (CF) is the most common lethal autosomal-recessive disorder in the Caucasian population.The pulmonary disease is the most relevant clinical expression of CF. Cystic fibrosis airways are under the effect of oxidants that are generated from increased inflammation and infections present in the lungs. Low glutathione (GSH) levels in epithelial lining fluid and neutrophils/blood plasma can impair antioxidant defences thus magnifying the oxidative stress in patients suffering from CF. On these basis increasing or restoring normal GSH levels has been proposed as a potentially useful strategy in CF. Cow's milk whey proteins because its high levels of cysteine and cystine, that are both precursors of endogenous GSH synthesis, can increase tissue levels of GSH as required in CF.Therefore the primary aim of this study was to evaluate the efficacy of a diet supplementation with a cow's milk whey protein formula (WPF) on oxidative stress in CF patients. The secondary goal was to assess the effects of this supplementation on clinical status and its tolerability. This is a prospective, open, clinical study conducted among CF patients in regular follow up at CF Centre of Genova, Italy in the period 2007-2010. Participants received whey proteins with WPF, which was administered orally once a day. This study deals with 59 patients.Mean follow up period was 18 months (range 3-36).Oxidative balance was evaluated as BAP test and d-ROMs test while improvement of respiratory function was assessed as FEV1% predicted for sex and age. In our data stress oxidative status showed to be favourably influenced byWPF administration as a matter of fact mean BAP value increased, even if in a not significantly way and,mainly, the d-ROMs value significantly decreased after WPF intake. Unfortunately stress oxidative condition in our data is not really associated with clinical status improvement. Conclusively we can say that, a nutraceutical formula, as WPF, has demonstrated a real effect on improvement oxidative stress in CF patients and is well accepted and well tolerated.
Article
Cystic fibrosis (CF) is a genetic disorder characterized by epithelial electrolyte transport abnormalities, elevated sweat Cl− concentrations, pancreatic insufficiency, and chronic lung disease in most patients. It is the most common potentially fatal genetic disorder in the Caucasian population, affecting 1 in 2400–3500 live births [1, 2]. It is an autosomal recessive disorder caused by a mutation in the gene for the cystic fibrosis transmembrane conductance regulator (CFTR), a membrane channel protein. The clinical significance of hepatobiliary disease in CF has not been well characterized primarily because of two factors: (1) pulmonary involvement leads to early mortality in a majority of patients, and (2) the clinical identification of CF-associated liver disease has been difficult because, although it is progressive, liver involvement is often asymptomatic until the appearance of end-stage complications. Recently, with improved pulmonary treatments, median life expectancy now exceeds 30 years [3] and CF-associated hepatobiliary disease is recognized and characterized more comprehensively. Liver disease is now the second major cause of death in CF [4]. In recent years, advances in our understanding of the function of CFTR in bile duct epithelia have provided a stronger scientific basis for the pathogenesis of the disease, leading to insights concerning potentially novel therapeutic approaches. The earliest reports of CF, probably date to the Middle Ages with reports of malnourished and “sickly” children that tasted “salty” when kissed [5].
Article
Significance: Cystic fibrosis (CF) is the most common lethal genetic disorder in the Caucasian people. It is due to the mutation of cystic fibrosis transmembrane conductance regulator (CFTR) gene located on the long arm of the chromosome 7, which encodes for CFTR protein. The latter, an adenosine triphosphate binding cassette, is a transmembrane chloride channel that is also involved in glutathione transport. As glutathione/glutathione disulfide constitutes the most important pool of cellular redox systems, CFTR defects could thus disrupt the intracellular redox balance. Resulting multisystemic diseases are essentially characterized by a chronic respiratory failure, a pancreatic insufficiency, an essential fatty acid deficiency (EFAD), and inadequate levels of antioxidant vitamins. Recent advances: The pathophysiology of CF is complex; however, several mechanisms are proposed, including oxidative stress (OxS) whose implication is recognized and has been clearly demonstrated in CF airways. Critical issues: Little is known about OxS intrinsic triggers and its own involvement in intestinal lipid disorders. Despite the regular administration of pancreatic supplements, high-fat high-calorie diets, and antioxidant fat-soluble vitamins, there is a persistence of steatorrhea, EFAD, and harmful OxS. Intriguingly, several trials with elevated doses of antioxidant vitamins have not yielded significant improvements. Future directions: The main sources and self-maintenance of OxS in CF should be clarified to improve treatment of patients. Therefore, this review will discuss the potential sources and study the mechanisms of OxS in the intestine, known to develop various complications, and its involvement in intestinal lipid disorders in CF patients.
Article
Full-text available
Vitamin C status and possible associations with the disease process in cystic fibrosis (CF) patients were investigated. Plasma vitamin C concentrations in patients from two different mid-European populations (Swiss, n = 62; Austrian, n = 60) taking no or low-dose vitamin C from multivitamin supplements did not differ from each other or from control subjects (n = 34). Vitamin C concentrations decreased with age (5.05 mumol.L-1, y-1). When followed up for 12 mo, patients had the highest plasma vitamin C concentrations in February and the lowest in May and August (P < 0.01); the decrease in vitamin C was accompanied by increases in plasma malondialdehyde (P < 0.001) and tumor necrosis factor alpha concentrations (P < 0.01). During supplementation with vitamin E for 2 mo or beta-carotene for 12 mo vitamin C concentrations did not change. They correlated inversely with white blood cell count (r = -0.36, P = 0.008), bands (r = -0.36, P = 0.02), alpha 1-acid glycoprotein (r = -0.45, P = 0.002), interleukin 6 (r = -0.46, P = 0.0006), and neutrophil elastase/alpha 1-proteinase inhibitor complexes (r = -0.34, P = 0.02). In patients with vitamin C concentrations < 40 mumol/L, all indexes of inflammation were relatively high, whereas those with concentrations > 80 mumol/L (upper quartile of control subjects) showed clearly lower values. These results are consistent with the hypothesis that by scavenging oxygen free radicals vitamin C interacts with an inflammation-amplifying cycle of activation of alveolar macrophages and neutrophils, release of proinflammatory cytokines and oxygen free radicals, and inactivation of antiproteases.
Article
Full-text available
The concentration of nitric oxide in exhaled air, a marker of airway inflammation, depends critically on the flow of exhalation. Therefore, the aim of this study was to determine the effect of varying the flow on end-expiratory NO concentration and NO output in children with asthma or cystic fibrosis (CF) and in healthy children.Nineteen children with stable asthma, 10 with CF, and 20 healthy children exhaled from TLC while controlling expiratory flow by means of a biofeedback signal at approximately 2, 5, 10 and 20% of their vital capacity per second. NO was measured in exhaled air with a chemiluminescence analyser. Comparisons between the three groups were made by analysing the NO concentration at the endexpiratory plateau and by calculating NO output at different flows.Exhaled NO decreased with increasing flow in all children. Children with asthma had significantly higher NO concentrations than healthy children, but only at the lowest flows. Asthmatics using inhaled steroids (n=13) tended to have lower median exhaled NO than those without steroids. The slope of linearized (log-log transformed) NO/flow plots was significantly steeper in asthmatics than in healthy controls. CF patients had a significantly lower NO concentration and output over the entire flow range studied, compared to asthmatic and control subjects, with a similar NO/flow slope as control subjects.In conclusion, the nitric oxide concentration in exhaled air is highly flow-dependent, and the nitric oxide-flow relationship differs between asthmatics versus cystic fibrosis patients and control subjects. Assessment of the nitric oxide/flow relationship may help in separating asthmatics from normal children.
Article
Oxidative stress induced by catecholamines is a well recognized toxic event. This effect has been extensively observed in the heart, where high levels of catecholamines cause enzyme inhibition, lipid peroxidation, energy depletion and myocardial necrosis. Catecholamines can be converted into o-quinones and undergo cyclization into aminochromes. This process can occur enzymati-cally or through autoxidation and involves the formation of free radicals. Aminochromes are highly reactive molecules that can cause oxidation of protein sulfhydryl groups and deamina-tion catalysis, among other deleterious effects; in addition, inhibition of some enzymes has been also reported. We have studied the effects of isoproterenol oxidation products (IOP) on glutathione reduc-tase (GR) activity in vitro. Isoproterenol (ISO) autoxidation was conducted at 37°C in the dark, for 4h at pH 7.0 and this process was monitored by UV spectrophotometry at both 340 and 490 nm. Addition of the autoxidized solution to GR in the presence of oxidized glutathione (GSSG) and NADPH showed that IOP inhibits GR in a competitive mode and that this effect increases during the 4h incubation period. This inhibitory effect of IOP was partially prevented by the addition of reduced glutathione (GSH), L-cysteine and ascorbic acid to the reaction mixtures.
Article
TNFα is implicated in several skeletal muscle pathologies including muscle wasting of cachexia. Muscle wasting and other conditions such as physical exercise and immobilization are also associated with disturbances in muscle glutathione status. Hence, it was of interest to investigate the role of endogenous glutathione status in TNFα induced NF-κB activation in skeletal muscle-derived cells. TNFα proved to be a potent inducer of transient NF-κB activation in L6 myoblasts. In buthioninesulfoximine (BSO) treated cells, TNFα induced NF-κB activation was markedly potentiated suggesting that such activation is sensitive to cellular GSH, but may have been independent of high levels of intracellular GSSG. Because this activation was inhibited by the antioxidant pyrrolidinedithiocarbamate (PDTC) the involvement of reactive oxygen species in this activation system seems likely. NF-κB activation in L6 cells was also observed in response to direct H2O2treatment. Results from GSSG reductase inhibited cells suggest that GSSG may participate in, but is not required for, TNFα induced NF-κB activation. The inhibitory effect of PDTC on NF-κB activation correlated with its effect on ICAM-1 expression suggesting that this GSH status modifying agent not only influenced nuclear translocation of NF-κB proteins but also regulated κB dependent transcription.
Article
Thanks to progress in zinc research, it is now possible to describe in more detail how zinc ions (Zn++) and nitrogen monoxide (NO), together with glutathione (GSH) and its oxidized form, GSSG, help to regulate immune responses to antigens. NO appears to be able to liberate Zn++ from metallothionein (MT), an intracellular storage molecule for metal ions such as zinc (Zn++) and copper (Cu++). Both Zn++ and Cu++ show a concentration-dependent inactivation of a protease essential for the proliferation of the AIDS virus HIV-1, while zinc can help prevent diabetes complications through its intracellular activation of the enzyme sorbitol dehydrogenase (SDH). A Zn++ deficiency can lead to a premature transition from efficient Th1-dependent cellular antiviral immune functions to Th2-dependent humoral immune functions. Deficiencies of Zn++, NO and/or GSH shift the Th1/Th2 balance towards Th2, as do deficiencies of any of the essential nutrients (ENs)- a group that includes methionine, cysteine, arginine, vitamins A, B, C and E, zinc and selenium (Se) - because these are necessary for the synthesis and maintenance of sufficient amounts of GSH, MT and NO.
Article
Earlier studies have reported the presence of inerleukin-1 (IL-1) and tumor necrosis factor (TNF) in the plasma of patients with cystic fibrosis (CF), but the results have been inconsistent. To investigate the relationships among plasma IL-1α, IL-1β, TNF, lipopolysaccharide (LPS), and clinical status, measurements were made before and after 14 days of intravenous antibiotic therapy in 13 patients with CF. In addition, whole blood cytokine production rates were measured in 18 hr cultures stimulated with 10 μg/mL LPS or sterile saline (control). On admission, patients with CF had significantly greater plasma levels of LPS and IL-1α compared with 20 healthy adult controls. In response to antibiotic therapy, the patients had statistically significant increases in weight, oxygen saturation, chest radiograph score, and forced expiratory volume in 1 second. They had significant decreases in pulse rate, residual volumeitotal lung capacity ratio, white blood count, neutrophil count, LPS concentration, and resting energy expenditure per kg body weight. There were no significant changes in the plasma concentrations of IL-1α, IL-1β, or TNF and no significant changes in the basal or stimulated whole blood production rates of IL-1α, IL-1β, or TNF. The immunological variables did not correlate significantly with clinical measurements of severity or the presence of fever. It is likely that in CF local pulmonary effects of cytokines are of more pathophysiologic significance than systemic effects. Pediatr Pulmonol. 1994; 18:21–27. © 1994 Wiley-Liss. Inc.
Article
Alterations of oxidants and antioxidants now appear to be pivotal in the development of bronchial hyperresponsiveness and bronchial asthma. To evaluate the potential protective role of the antioxidant reduced glutathione (GSH) administered by ultrasonic nebulizer on metacholine-induced bronchoconstriction, we designed a double-blind, randomized study enrolling 18 subjects with mild asthma and previous bronchoconstriction after a methacholine challenge; we did not find a statistically significant decrease in bronchoconstriction after premedication with inhaled GSH. Further investigation under different experimental conditions is warranted because our information about the mode of action and pharmacokinetics of GSH is still incomplete and sometimes the data are conflicting.
Article
Secretory leukoprotease inhibitor (SLPI) and alpha(1)-protease inhibitor (alpha(1)-PI) are powerful antiproteases currently under investigation for their potential to protect the lung from neutrophil elastase (NE). The aim of this study was to determine whether the recombinant form of SLPI (rSLPI) and alpha(1)-PI show different grades of loss of inhibitory activity when exposed to reactive oxygen metabolites. We incubated rSLPI and alpha(1)-PI with N-chlorosuccinimide (NCS), chloramines, activated polymorphonuclear leucocytes (PMNs) and activated alveolar macrophages (AMs). Under all conditions evaluated, both antiproteases were partially inactivated, The resulting anti-NE activity of rSLPI was not significantly different from that of alpha(1)-PI after exposure to NCS (p>0.5), chloramines (p>0.6), activated PMNs (p> 0.07) and activated AMs (p>0.9). In conclusion, recombinant secretory leukoprotease inhibitor and alpha(1)-protease inhibitor lose antineutrophil elastase activity to a similar extent when expressed to conditions that may be present in inflammatory lung disorders.
Article
The inducible nitric oxide synthase (iNOS) promoter contains nuclear factor κB (NF-κB) binding sites. NF-κB activation is determined, in part, by the intracellular redox status. The aim of this study was to determine the importance of the cellular glutathione status in relation to NF-κB activation and iNOS expression in hepatocytes in vivo and in vitro. For in vivo experiments, rats were injected with endotoxin and sacrificed 6 hours later. Glutathione was depleted by diethylmaleate. For in vitro experiments, cultured hepatocytes from untreated rats were exposed to a cytokine mixture. Glutathione levels were depleted by diethylmaleate and restored by N-acetylcysteine. iNOS expression was assessed by Western blot, reverse transcription polymerase chain reaction, nitric oxide (NO) metabolites, and immunohistochemistry. NF-κB binding was assessed by electrophoretic mobility shift assay. Endotoxin-induced iNOS expression in rat liver was prominent in hepatocytes, Kupffer cells, and inflammatory cells, in particular neutrophils. Glutathione depletion prevented iNOS induction in hepatocytes, but not in inflammatory cells. iNOS protein levels were in accordance with iNOS messenger RNA and NO metabolites in plasma. Glutathione depletion did not affect neutrophil infiltration. Cytokines strongly induced iNOS in cultured hepatocytes. Induction was prevented by glutathione depletion and could be restored by addition of N-acetylcysteine. NF-κB binding correlated with iNOS induction. In conclusion, in this study we show that iNOS induction in hepatocytes in vivo and in vitro is dependent on the intracellular glutathione status and correlates with NF-κB binding. Glutathione-depletion has no effect on the expression of iNOS in inflammatory cells, nor on neutrophil infiltration
Article
The ability of nonprotein thiols to modulate rates of protein synthesis was investigated in isolated rat hepatocytes. Addition of cysteine stimulates protein labelling by [14C] Leucine. Glutahione depletion, induced by in vivod administration of L-buthionine sulfoximine and diethylmaleate, did not alter the effect of cysteine, although it decreased the rate of protein synthesis by 32%. The effect of cysteine on protein synthesis does not seem to be related to a perturbatin of the redox state of the NAD+/NADH system or to changes in the rate of gluconeogenic pathway. The following observations indicate that cysteine may stimulate protein syntheis by increasing intracellular levels of aspartate: 1. Amino-oxyacetate, an inhibitor of pyridoxyal-dependent enzymes, inhibits protein labelling and decreases aspartate cellular content, whereas most amino acids accumulate or remain unchanged; 2. Cysteine, in the absence or in the presence of amino-ocycetate, stimulates protein labelling and induces aspartate accumulation, although mot amino acids diminish or remain unchanged.