Download as pdf or txt
Download as pdf or txt
You are on page 1of 27

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/5690064

The central role of glutathione in the pathophysiology of human disease

Article  in  Archives of Physiology and Biochemistry · October 2007


DOI: 10.1080/13813450701661198 · Source: PubMed

CITATIONS READS
334 12,656

4 authors, including:

Rodrigo Franco Aglaia Pappa


University of Nebraska at Lincoln Democritus University of Thrace
107 PUBLICATIONS   9,914 CITATIONS    101 PUBLICATIONS   3,746 CITATIONS   

SEE PROFILE SEE PROFILE

Mihalis I Panayiotidis
Northumbria University
92 PUBLICATIONS   3,852 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Guest editing a special Issue for Antioxidant titled as: "Natural Products in Health Promotion and Disease Prevention" View project

Redox Metabolism in Parkinson´s Disease View project

All content following this page was uploaded by Mihalis I Panayiotidis on 14 July 2014.

The user has requested enhancement of the downloaded file.


This article was downloaded by:[University of Nevada Reno]
On: 21 April 2008
Access Details: [subscription number 791859321]
Publisher: Informa Healthcare
Informa Ltd Registered in England and Wales Registered Number: 1072954
Registered office: Mortimer House, 37-41 Mortimer Street, London W1T 3JH, UK

Archives Of Physiology And


Biochemistry
Formerly Archives Internationales de
Physiologie, de Biochimie et de Biophysique,
founded in 1904
Publication details, including instructions for authors and subscription information:
http://www.informaworld.com/smpp/title~content=t713817673
The central role of glutathione in the pathophysiology of
human diseases
R. Franco a; O. J. Schoneveld a; A. Pappa b; M. I. Panayiotidis c
a
Laboratory of Signal Transduction, National Institute of Environmental Health
Sciences, National Institutes of Health, Research Triangle Park, NC
b
Department of Molecular Biology and Genetics, Democritus University of Thrace,
Alexandroupolis, Greece
c
Environmental & Occupational Health, School of Public Health, University of Nevada at Reno, Reno, NV

Online Publication Date: 01 October 2007


To cite this Article: Franco, R., Schoneveld, O. J., Pappa, A. and Panayiotidis, M. I. (2007) 'The central role of
glutathione in the pathophysiology of human diseases', Archives Of Physiology And Biochemistry, 113:4, 234 - 258
To link to this article: DOI: 10.1080/13813450701661198
URL: http://dx.doi.org/10.1080/13813450701661198

PLEASE SCROLL DOWN FOR ARTICLE

Full terms and conditions of use: http://www.informaworld.com/terms-and-conditions-of-access.pdf


This article maybe used for research, teaching and private study purposes. Any substantial or systematic reproduction,
re-distribution, re-selling, loan or sub-licensing, systematic supply or distribution in any form to anyone is expressly
forbidden.
The publisher does not give any warranty express or implied or make any representation that the contents will be
complete or accurate or up to date. The accuracy of any instructions, formulae and drug doses should be
independently verified with primary sources. The publisher shall not be liable for any loss, actions, claims, proceedings,
demand or costs or damages whatsoever or howsoever caused arising directly or indirectly in connection with or
arising out of the use of this material.
Archives of Physiology and Biochemistry, October/December 2007; 113(4/5): 234 – 258
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

REVIEW ARTICLE

The central role of glutathione in the pathophysiology of human diseases

R. FRANCO1*, O. J. SCHONEVELD1, A. PAPPA2, & M. I. PANAYIOTIDIS3


1
Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health,
Research Triangle Park, NC 27709, 2Department of Molecular Biology and Genetics, Democritus University of Thrace,
Dimitras 19, 68100 Alexandroupolis, Greece and 3Environmental & Occupational Health, School of Public Health, University
of Nevada at Reno, Reno, NV

Abstract
Reduced glutathione (L-g-glutamyl-L-cysteinyl-glycine, GSH) is the prevalent low-molecular-weight thiol in mammalian
cells. It is formed in a two-step enzymatic process including, first, the formation of g-glutamylcysteine from glutamate and
cysteine, by the activity of the g-glutamylcysteine synthetase; and second, the formation of GSH by the activity of GSH
sythetase which uses g-glutamylcysteine and glycine as substrates. While its synthesis and metabolism occur intracellularly, its
catabolism occurs extracellularly by a series of enzymatic and plasma membrane transport steps. Glutathione metabolism and
transport participates in many cellular reactions including: antioxidant defense of the cell, drug detoxification and cell
signaling (involved in the regulation of gene expression, apoptosis and cell proliferation). Alterations in its concentration have
also been demonstrated to be a common feature of many pathological conditions including diabetes, cancer, AIDS,
neurodegenerative and liver diseases. Additionally, GSH catabolism has been recently reported to modulate redox-sensitive
components of signal transduction cascades. In this manuscript, we review the current state of knowledge on the role of GSH
in the pathogenesis of human diseases with the aim to underscore its relevance in translational research for future therapeutic
treatment design.

Key words: GSH, apoptosis, human diseases, human disorders, ROS, free radicals, oxidative stress, thiols, glutathionylation,
glutathiolation, glutathione depletion, glutathione S-transferases, glutathione peroxidase, glutathione reductase, nitrosylation.
.
Abbreviations: GSH, reduced glutathione; GSSG, glutathione disulfide; GS , thyil radical; ROS, reactive oxygen
species; RNS, reactive nitrogen species; RS, reactive species; g-GCS, g-glutamylcysteine synthetase; GS, glutathione
synthetase (synthase); g-GT, g-glutamyl transpeptidase; GPX, glutathione peroxidase; GR, glutathione reductase;
GST, glutathione S-transferase; GSNO, nitrosoglutathione; OATP, organic anion transporting polypeptide; MRP,
multidrug resistance proteins; OA7, organic anions; NAC, N-acetyl-L-cysteine; NO, nitric oxide; OH, hydroxyl
.

radical; H2O2, hydrogen peroxide; O27, superoxide anion; ONOO7, peroxynitrite.


.

properties grow exponentially. Today it is widely


Introduction
accepted that GSH not only acts as a reducing agent
Thiol containing compounds are transcendental in and a major antioxidant within the cells maintaining
many biochemical and pharmacological reactions a tight control of the redox status, but it also acts as
(Dickinson & Forman, 2002). In its reduced form, mediator of many other physiological reactions
glutathione (L-g-glutamyl-L-cysteinyl-glycine, GSH) including cellular signaling (involved in cell cycle
is the most abundant non-protein thiol in mamma- regulation, proliferation and apoptosis), metabolism
lian cells and the prevalent low-molecular weight of xenobiotics, thiol disulfide exchange reactions,
peptide in eukaryotic cells (Figure 1). It was initially and also as an important reservoir of cysteine. It is
described as a potent reducing agent; however, many not surprising then that a large body of bibliography
other cellular functions have been recently ascribed is available on the role of GSH in several
to GSH, making the scientific interest in its biological human diseases including cancer, neurodegenerative

Correspondence: Dr. R. Franco, Laboratory of Signal Transduction, National Institute of Environmental Health Sciences (NIEHS), National Institutes of
Health (NIH), PO Box 12233, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709. Tel: (919) 541-5387. Fax: (919) 541-1898. E-mail:
franco2@mail.nih.gov, *Unidad de Biomedicina, Facultad de Estudios Superiores-Istacala, Universidad Nacional Autónoma de México, Tlalnepantla, México,
México. E-mail: rfranco&campus.iztacala.unam.mx and Dr. M. I. Panayiotidis, Environmental & Occupational Health, School of Public Health, University of
Nevada at Reno, Reno, NV. Tel: 775-682-7082. Fax: 775-784-1340. E-mail: panayiotidism@unc.edu

Received for publication 16 March 2007. Accepted 20 July 2007.


ISSN 1381-3455 print/ISSN 1744-4160 online ª 2007 Informa UK Ltd.
DOI: 10.1080/13813450701661198
GSH in human health and disease 235

different organelles including mitochondria, peroxi-


Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

somes, nuclear matrix and endoplasmic reticulum


after its cytosolic synthesis (Valencia et al., 2001b;
Valencia et al., 2001d; Jefferies et al., 2003; Wu et al.,
2004).
The intracellular concentration of GSH reflects a
dynamic balance between synthesis, consumption
rate (metabolism), and its transport. Glutathione
synthesis starts by the formation of g-glutamylcysteine
synthetase, a dipeptide form by the combination
of glutamate and cysteine catalyzed by the
g-glutamylcysteine synthetase (g-GCS), also known
as g-glutamylcysteine ligase (Figure 2). In this
reaction, the g-carboxyl group of glutamate reacts
with the amino group of cysteine to form a peptidic
g-linkage that protects GSH from hydrolysis by
Figure 1. Structure of reduced glutathione (GSH). Glutathione (L-g- intracellular peptidases. After this initial step, glycine
glutamyl-L-cysteinyl-glycine, GSH) is a linear tripeptide formed is added to g-glutamylcysteine by the activity of GSH
from the amino acids glycine, cysteine and glutamate (MW sythetase (GS). Both enzymatic steps consume one
307.4 g mol71). The peptidic g-linkage between glutamate and molecule of ATP per catalytic cycle. The presence of
cysteine protects GSH from hydrolysis by intracellular peptidases,
the C-terminal glycine protects it against cleavage by
while the presence of the C-terminal glycine protects it against
cleavage by intracellular g-glutamylcyclotransferases. The cysteinyl intracellular g-glutamylcyclotransferases. Glutathione
moiety of GSH provides the reactive thiol group (-SH group) that acts as a feedback inhibitor of its synthesis since it can
allows GSH to participate in a wide variety of metabolic processes competitively inhibit the g-GCS activity. On the other
including oxidation-reduction (redox) reactions and nucleophilic hand, the availability of cysteine is the rate-limiting
displacement or addition-type reactions.
factor in GSH formation (Sies, 1999; Dickinson &
Forman, 2002; Wu et al., 2004; Estrela et al., 2006).
diseases (Alzheimer, Parkinson), acquired immune Glutathione can also be synthesized through sal-
deficiency syndrome (AIDS), aging, cystic fibrosis, vage pathways that involve its catabolism (Figure 3),
liver and heart disease, ischemia, stroke, seizure, or through its recycling after its oxidation (this is
sickle cell anemia and metabolic diseases (diabetes discussed in the next section). Glutathione catabo-
and obesity) (Sies, 1999; Reid & Jahoor, 2001; lism occurs extracellularly by the activity of the
Jefferies et al., 2003; Pompella et al., 2003; Town- g-glutamyl transpeptidase (g-GT). The g-GT is
send et al., 2003; Dalton et al., 2004; Wu et al., expressed mainly on the apical surface of cells and
2004). The aim of this review is to summarize and initiates the catabolism of not only GSH, but of
highlight the molecular events that regulate the glutathione S-conjugates and glutathione-complexes.
pathogenesis of distinct human diseases by altera- g-Glutamyl transpeptidase removes the g-glutamyl
tions in GSH metabolism, transport and catabolism. moiety from GSH and GSH-conjugated compounds
by transferring it to other acceptors (amino acids or
other dipeptides), producing cysteinylglycine or
Glutathione synthesis, transport and
cysteinylglycine-conjugates. These products are
catabolism
further hydrolyzed by ectoprotein dipeptidases
Glutathione is a ubiquitous molecule that is pro- which remove the peptide bond between
duced intracellularly in all organs and cell types, but cysteine and glycine. Cysteine, together with the
it is more abundant in liver and lung tissues. It is a g-glutamyl-amino acids formed from the action of the
linear tripeptide formed from the amino acids g-GT, are then uptaken by the activity of specific
glycine, cysteine and glutamate (M.W. 307.4 g transporters. The g-glutamyl-derivatives are further
mol71) (Figure 1), attaining concentrations of 1 to substrates for the g-glutamyl cyclotransferase that
10 mM in many different cell types. Plasma GSH forms 5-oxoproline and the corresponding amino
concentrations on the other hand are relatively low acid. 5-Oxoproline is finally converted to glutamate
*0.01 mM, mainly because of its rapid catabolism. by the activity of the 5-oxoprolinase. On the other
Intracellular GSH can exist as a monomer in its hand, S-cysteine conjugates, as a result of GSH
reduced form, or as a disulfide dimmer formed due S-conjugates breakdown by g-GT, are acetylated on
to its oxidation (GSSG) which usually accounts for the amino group of the cysteinyl residue by intracel-
less than 1% of the total intracellular glutathione lular N-acetylfransferases to form mercapturic acids
content. Additionally, an important fraction of total (Paolicchi et al., 2002; Estrela et al., 2006).
intracellular GSH can be found as thioester, mercap- Some studies have suggested the existence of a
tide or other thioester forms (GSH conjugates). Naþ-coupled GSH transport influx based on the idea
Glutathione is 85 to 90% freely distributed in the that rates of intracellular GSH synthesis and efflux
cytosol, although it can be compartmentalized in across the plasma membrane may not account by
236 R. Franco et al.
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

Figure 2. Overview of de novo glutathione synthesis. Reduced glutathione or GSH is a tripeptide formed by the amino acids glutamate, cysteine
and glycine, whose availability in the cell depends on cellular uptake by amino acid transporters or on other biochemical pathways.
Glutathione synthesis is mediated by the activity of the g-glutamylcysteine synthetase (g-GCT) and the glutathione synthetase (GS).
Glutathione is freely distributed in the cytosol, reaching a concentration of up to 10 mM in animal cells, and it can be compartmentalized (up
to 15%) to some degree in mitochondria, endoplasmic reticulum or nuclei matrix. Both cytosolic and compartmentalized GSH have been
reported to participate in distinct biological processes.

Figure 3. Glutathione salvage pathway. Glutathione can also be synthesized through salvage pathways that involve its catabolism. Glutathione
catabolism is mediated by the g-glutamyl transpeptidase (g-GT) expressed mainly on the apical surface of cells. It mediates the catabolism of
not only GSH, but of GSSG and GSH-conjugates. g-Glutamyl transpeptidase removes the g-glutamyl moiety from GSH and GSH-
conjugated compounds by its transfer to other acceptors (amino acids, aa, or other dipeptides), thus producing cysteinylglycine (Cys-Gly) or
cys-gly-conjugates, and g-glutamyl-amino acid (g-Glu-aa) products. These products are further hydrolyzed by ectoprotein dipeptidases
(DPT) which remove the peptide bond between cysteine and glycine. The g-glut-aa, glycine and cysteine (or cys-conjugates) formed from the
action of the g-GT are then uptaken by the activity of specific transporters. The g-glutamyl-derivatives are further substrates for the
g-glutamyl cyclotransferase (g-GCT) which forms 5-oxoproline and the corresponding amino acid. 5-Oxoproline is finally converted to
glutamate by the activity of the 5-oxoprolinase (OXP). On the other hand, S-cysteine conjugates, as a result of GSH S-conjugates breakdown
by g-GT, are acetylated on the amino group of the cysteinyl residue by intracellular N-acetylfransferases (NAT) to form mercapturic acids.
GSH in human health and disease 237

themselves for the maintenance of relatively high protein (MRP, encoded by ABCC genes) that is a
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

intracellular GSH concentrations. However, its ex- subfamily of the ATP binding cassette transporters
istence is still controversial (Gukasyan et al., 2006). (ABC transporters), and the organic anion transport
In the basolateral membrane of renal proximal polypeptide proteins (OATP, encoded by SLCO
tubular cells, the existence of two different GSH genes) (Hammond et al., 2001; Ballatori et al., 2005).
influx transporters has been proposed. The first one The MRP transporters have been demonstrated to
involves a Naþ-independent mechanism which has act as cotransporters of organic anions (OA7) and
been proposed to be mediated by the organic anion GSH. In addition, they also act as transporters of
transporters 1 and 3 (OAT1/3) that act as exchangers GSH-conjugated xenobiotics and GSH-conjugated
of GSH for 2-oxoglutarate. On the other hand a metabolites (e.g. the cysteinyl leukotriene C4) that
second Naþ-dependent influx of GSH has been attri- must be exported from the cells in which they are
buted to the sodium dicarboxylate cotransporter-2 formed to avoid deleterious effects. This efflux
(SDCT-2) (Lash, 2005). However, the biological confers drug resistance to tumor cells and can
importance of these transporters in GSH uptake in protect normal cells from toxic insults. Multidrug
other tissues and cell lines remains to be explored. resistance proteins also export GSSG thus suggesting
Because GSH degradation (catabolism) occurs a role of MRP proteins in cellular responses to
extracellularly, the export of GSH, GSSG and oxidative stress. In any case, MRP transport of
GSH-adducts is an important step in its turnover. organic anions including drugs and conjugated OA7
Relatively little is known at the molecular level about requires the presence of GSH and the hydrolysis of
GSH efflux transporters and pumps (Figure 4). To ATP (Ballatori et al., 2005; Cole & Deeley, 2006).
date two GSH transporter families have been well On the other hand the OATP transporters were
implicated in GSH efflux across the plasma mem- initially proposed to act as exchangers of GSH/OA7.
brane. These include the multidrug resistance In this way, GSH efflux is stimulated by the presence

Figure 4. Glutathione transporters. Plasma membrane GSH efflux pumps. Two GSH transporter families have been implicated in GSH efflux
across the plasma membrane. The first group includes the multidrug resistant protein family (MRP) which acts as cotransporters of GSH
coupled to the extrusion of an OA7 (organic anion) and requires the use of energy. MRP proteins have also proposed to transport GSSG and
GSH-conjugates. The second group includes the organic anion transporting polipeptide proteins OATP which act as GSH/OA- exchanger.
This exchange is stimulated by the presence of high extracellular concentrations of OA7 and is driven by the electrochemical gradient of
GSH across the plasma membrane. Other proposed candidates for GSH efflux are hemichannels (connexins, CX), CFTR and RLIP76.
Mitochondrial GSH transporters. The charged nature of GSH (negative at physiological pH) suggests that GSH cannot passively diffuse into
the mitochondria, because the mitochondrial matrix has a negative potential relative to the cytoplasm. Some of the strongest candidates
involved in mitochondrial import of cytosolic GSH include the dicarboxylate transporter (DIC) and the 2-oxoglutarate transporter (OGC)
which act as exchangers of Pi2þ (inorganic phosphorous) and 2-oxoglutarate (2-OG27) for cytosolic GSH. This transport dissipates any
concentration gradient between cytoplasm and mitochondrial whose concentrations are both *5 mM. Additionally, mitochondrial
GSH transport allows this pool to be relatively independent from the cytosol, which when depleted does not affect the mitochondrial
GSH pool.
238 R. Franco et al.

of a wide range of structurally unrelated OA7


Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

Distinct biological functions of GSH


substrates (trans-stimulation) demonstrating the
metabolism
wide inespecificity of the OA7 binding site in the
OATP proteins. Glutathione transport by OATP The cysteinyl moiety of GSH provides the reactive
proteins is driven by the outwardly directed electro- thiol group (-SH group) that allows GSH to
chemical gradient across the plasma membrane, thus participate in a wide variety of metabolic processes
it can be reversed by increases in the extracellular including oxidation-reduction (redox) reactions and
GSH concentration demonstrating its potential nucleophilic displacement or addition-type reactions.
bidirectionality. As mentioned earlier, GSH is pre- Glutathione reacts widely to form a number of
sent in high concentrations within the cells (10 mM), different metabolites. These reactions can be divided
whereas blood plasma concentrations are three into those involving principally the g-glutamyl por-
orders of magnitude lower (approximately tion of the tripeptide such as in the case of the
0.01 mM). Because GSH is negatively charged at reactions mediated by g-glutamyl transpeptidases,
physiological pH, there is also a large negative and those involving the sulfhydryl moiety that
intracellular potential (730 to 760 mV) that facil- include oxidation-reduction and nucleophilic reac-
itates its extrusion from the cell. In this way, the tions (Meister, 1995a; Meister, 1995b; Jefferies et al.,
combined chemical and electrical GSH gradients 2003; Wu et al., 2004).
serve as a powerful driving force for cellular uptake of
solutes by OATP (Hagenbuch & Meier, 2004;
Antioxidant and redox balance regulator
Ballatori et al., 2005). Recently, however, some
studies have challenged the view of OATP transpor- Glutathione is a strong reducing agent that con-
ters as GSH/OA7 exchangers. Bi-directional GSH/ tributes to key antioxidant metabolic pathways by
OA7 has been reported in different cell types acting either as a proton donor, or as a cofactor of
including human cell lines (Garcia-Ruiz et al., nucleophilic conjugates. Because GSH constitutes
1992; Sze et al., 1993; Iantomasi et al., 1997; Li the major intracellular antioxidant defense within the
et al., 1998; Li et al., 2000; Lee et al., 2001; Ng et al., cells, its depletion might be either a cause or a
2003). Although OATP (human)/Oatp (rat, mouse) prerequisite for the generation of ROS which could
proteins were initially reported to mediate this potentially mediate a wide range of signaling events
exchange transport (Li et al., 1998; Li et al., 2000; (Figure 5, Table I). In this way, GSH has been
Hagenbuch & Meier, 2004), recent studies have shown to scavenge a wide range of reactive species
suggested that GSH/OA7 exchange is not mediated (RS) including reactive oxygen species (ROS) and
by this family of transporters (Briz et al., 2006; reactive nitrogen species (RNS). In this way, GSH
Mahagita et al., 2007). Thus there is a strong can scavenge superoxide anion ( O .
2 ), hydroxyl
possibility that GSH/OA7 is mediated by a different radical ( OH7), and singlet oxygen (1O2) directly,
.

and still uncharacterized entity. by donating electrons and becoming oxidized to thyil
.
Other less characterized pathways for GSH and radical (GS ). It is important to mention that the
GSH-conjugates efflux transport are those involving reductive ability of GSH, is associated with the
.
the cystic fibrosis transmembrane conductance formation of thiyl radical (GS ) which if not efficiently
regulator (CFTR), the Ral-regulated effector pro- removed (by ascorbate for example), can lead to pro-
tein (RLIP76), and hemichannels (connexins). oxidant reactions and peroxidative injury (Wlodek,
.
CFTR (ABCC7) is a member of the ABCC/MRP 2002). Two GS species can react further to form a
family of transporters which in addition to its GSSG molecule. In general, RS are reduced or
function as a chloride channel, has also been inactivated through the generation of disulfide bonds
suggested to mediate either direct transport of between two glutathione molecules to form GSSG.
GSH or to modulate GSH transport by other Moreover, GSH can directly scavenge protein- and
proteins (Hudson, 2001). RLIP76 (RALBP1) is a DNA-radicals. Additionally, non-enzymatic reaction
76 kDa Ral-binding, Rho/Rac-GAP and Ral effector of nitric oxide (NO) with GSH convey to the
protein which was recently proposed to be a novel formation of the relatively stable S-nitrosoglutathione
multispecific transporter of xenobiotics as well as (GSNO), which can be inactivated by its conjugation
GSH-conjugates with inherent ATPase activity to proteins through S-nitrosylation reactions. Glu-
(Awasthi et al., 2003). Finally, connexins are a tathione can also catalytically detoxify cells from
large family of transmembrane proteins that, when hydroperoxides (H2O2), peroxynitrite (OONO7)
apposed to other cells’ connexins, form functional and lipid peroxides by the action of GSH peroxidases
intercellular channels or gap junctions; and when (GPXs), peroxiredoxins (PXRs), and phospholipid
unapposed, form hemichannels. They control the hydroperoxide GSH peroxidases (PHGPX) (Mates,
diffusion driven passage of ions and molecules of a 2000; Valko et al., 2007). Glutathione peroxidases
molecular mass of up to 1 kDa. Recently, hemi- are selenoproteins that vary in their peroxide
channels have been suggested to mediate the efflux substrate specificity. On the other hand, peroxire-
of glutathione in astrocytes (Contreras et al., 2004; doxins catalyze the reduction of H2O2 by thiols
Rana & Dringen, 2006). including GSH using cysteine in its thiolate (S7)
GSH in human health and disease 239
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

Figure 5. Metabolism of GSH. GSH metabolism can be arbitrarily classified in three different types of functions including antioxidant, as a
mediator of thiol/exchange reactions, and as a factor in nucleophile conjugation. As a strong reducing agent, GSH is involved in free radical
. .
or reactive species (RS) and metal oxido-reduction reactions that lead to the formation of the thyil radical GS . Two GS species can further
react and form a GSSG molecule (1). Additionally, GSH can catalytically detoxify cells from peroxides (PX) by the action of GSH
peroxidases (GPXs) peroxiredoxins (PXRs) and phospholipid hydroperoxide GSH peroxidases (PHGPX). Glutathione disulfide (GSSG) is
further reduced to GSH by NADPH through glutathione reductase (GR) (2). The antioxidant role of GSH is also mediated by its role in
other primary antioxidant systems such as that of the dehydroascorbate reductase (DHR) which regenerates oxidized ascorbate
(dehydroascorbate) (3). Glutathione also participates in thiol/exchange reactions (S-glutathionylation and S-nitrosylation) as a reversible
mechanism for post-translational modification of proteins. The main reactions involved in this phenomenon are protein-SH/GSSG exchange
reactions and NO mediated formation of S-nitrosoglutathione (GSNO) and which mediates S-nitrosylation of proteins. Disulfide linkages
are mainly removed (dethiolation or deglutathionylation) by the activity of enzymes such as glutaredoxin (GRX) or thioredoxin (TRX) (4).
Finally, as a nucleophile, GSH can react with a wide variety of electrophiles (including xenobiotics and endogenous metabolites) in a reaction
catalyzed by glutathione transferases (GST) thus leading to the formation of GSH-conjugates or adducts (5).

form in their active site rather than selenium. increases in GSSG are counteracted by its rapid
Furthermore, H2O2 can spontaneously react with reduction by GR (Forman et al., 2002). However,
S7 to form sulfenic acid. Since these S7 molecules severe oxidative stress impairs GR activity leading to
are often part of active sites of a variety of proteins increased GSSG accumulation in the cell which in
including protein tyrosine phoshatases, alteration of most cases can be actively extruded through specific
protein function by H2O2 can be modulated by GSH. transporters. The reducing power of ascorbate also
Other enzymes including GSH-transferases (GST) helps to maintain systemic GSH homeostasis
have been shown to help in the metabolism of RS like (Gutierrez-Correa & Stoppani, 1997; Vander Jagt
NO and peroxides. Finally, GSH also participates in et al., 1997). Glutathione reductase has been
the antioxidant defense against carbon-centered reported regulated at the expression level by several
radicals. (Mates, 2000; Valko et al., 2007). oxidative stress conditions (Masella et al., 2005).
Glutathione disulfide formed in the process of RS Recently the activity of both GR and GPX have been
scavenging, also known as oxidized GSH, is reduced shown to be important predictors of the general
to glutathione by the action of glutathione reductase tissue redox state (Yang et al., 2006a).
(GR) (Wu et al., 2004) (Figure 5). Glutathione The antioxidant function of GSH is also mediated
reductase is a flavoenzyme represented as a single- by its role in other primary antioxidant systems.
copy gene in humans. It requires reduced nicotina- Several GSH-dependent enzymes have been de-
mide adenine dinucleotide phosphate (NADPH) as scribed to have dehydroascorbate reductase activity.
an electron donor reductant, which is maintained In addition, novel dehydroascorbate reductases
predominantly by the pentose phosphate shunt. (DHR) have been reported to act as GSH-dependent
Under physiological conditions GSSG is maintained enzymes. In this case, DHR activity is provided by
at *1% of total glutathione concentration and the electron donor capacity of GSH that allows the
240 R. Franco et al.

regeneration of oxidized ascorbate (dehydroascor- form. Thus, changes in the GSH/GSSG equilibrium
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

bate) (Meister, 1994; Maellaro et al., 1997; Masella within the cell may regulate certain metabolic path-
et al., 2005). Glutathione may also aid in the storage ways by activating or inactivating key enzymes.
and transfer of cysteine because of its reducing Without a process to reduce protein disulfides,
properties. Cysteine’s rapid autoxidation to cystine vulnerable cysteinyl residues of essential enzymes
produces potentially toxic oxygen radicals and so might remain oxidized, leading to changes in their
most of the non-protein cysteine is stored as GSH. catalytic activity (see section below). Higher-order
Thus, the ability of many cells to transport GSH into thiol cell systems like metallothioneins and thior-
the extracellular compartment may help in the edoxins are regulated by the GSH/GSSG balance
transport of cysteine and might also protect the cell (Schafer & Buettner, 2001; Jones, 2006).
membrane by reducing harmful reactive compounds
such as RS (Meister, 1995a; Meister, 1995b; Jefferies
Thiol/exchange reactions. Glutathionylation
et al., 2003; Wu et al., 2004).
(glutathyolation) of proteins
Changes in the intracellular thiol-disulfide (GSH/
GSSG) balance within the cell can be used as an Several proteins involved in cellular signaling have
indicator of the redox status of the cell. As mentioned critical thiols whose function can be altered by redox
above, under conditions of oxidative stress, GSH is modifications, including receptors, kinases, tran-
consumed by scavenging free radicals while GSSG scription factors and proteins involved in ubiquitina-
levels increase. The ratio of [GSH/GSSG] and the tion. Proteins with sulhydryls 7SH (thiol groups)
reduction potential (mV) are two ways to express can respond to oxidative stress by forming disulfides.
the intracellular redox environment of the cell. The The formation of disulfide bonds within a protein
GSH/GSSG couple is considered to be a cellular (intra) or between two proteins (inter) are reversed
redox buffer because it provides a very large pool of by protein disulfide isomerases. On the other hand,
reducing equivalents. Whereas many proteins are the formation of mixed disulfides with low-molecular
active when the key sulfhydryls are in the thiol form, mass thiols such as cysteine, homocysteine and GSH
others require them to be in the oxidized disulfide (s-thiolation) generates mixed protein/non-protein
disulfides (s-thiolated proteins). Because GSH is the
most available substrate in these reactions most of the
Table I. Examples of GSH-mediated oxido-reduction reactions. At S-thiolation processes involve S-glutathionylation of
physiological pH (7.4) and in solution, GSH becomes dissociated proteins. The importance of GSH in mixed disulfide
to GS7 (thiolate anion) þ Hþ. Thus, GSH in its GS7 form
mediates most of the reactions described below.
formation is demonstrated by the observation that
nearly 85% of mixed protein disulfides contain GSH.
Precursors Products Potential modes for S-glutathionylation modification
of protein thiols are exemplified in Table II. The
GSH þ OH7
. .
GS þ H2O
GSH þ O
.
þ GS þ H2O
2 þH
GSH þ NO þ O2 GSNO þ O2
. .
GSH þ R GS þ RH Table II. Potential modes for GSH mediated S-glutathionylation
GSH þ DNA
. .
GS þ DNA and S-nitrosylation.
2GSH þ H2O2 GSSG þ 2H2O
S-Glutathionylation
2GSH þ LOOH GSSG þ LOH þ H2O
Precursors Products
2GSH GSSG þ Ascorbic acid
þ Dihydroascorbate GSSG þ Protein-SH Protein-SSG þ GSH
2GSH þ Protein-SSX GSSG þ Protein-(SH2)X GSH þ Protein-SOH Protein-SSG þ H2O
. .
GS þ Protein-S Protein-SSG
GSH þ Protein-SSG GSSG þ Protein-SH
GS-OH þ Protein-SH Protein-SSG þ H2O
GSH þ Mn Mn71 þ GS
.
.
GSNO þ Protein-S Protein-SSG þ NO
GSH þ PGG2 GSSG þ PGH2 GSNO þ Protein-S7 Protein-SSG þ NO7
.
GS þ GS
.
GSSG GSH þ Protein-SNO Protein-SSG þ NO
Protein  SSG þ O2
.

GSSG þ NADPH þ Hþ 2GSH þ NADPþ GSH þ Protein-S þ O2


GS þ Protein-S7 þ O2 Protein  SSG þ O2
.

Abbreviations: GSH, reduced glutathione; GSSG, glutathione S-Nitrosylation


.
disulfide; GS , thyil radical; GPX, glutathione peroxidase; GR,
glutathione reductase; GSNO, nitrosoglutathione; NO, nitric GSNO þ Protein-SH Protein-SNO þ GSH
oxide; OH, hydroxyl radical; H2O2, hydrogen peroxide; O27,
. .

superoxide anion; NADPH, b-nicotinamide adenine dinucleotide Abbreviations: GSH, reduced glutathione; GSSG, glutathione
.
phosphate; Protein-SSG; protein mixed disulfide; Protein-SSX, disulfide; GS , thyil radical; GSNO, nitrosoglutathione; NO, nitric
oxide; O27, superoxide anion; Protein-SSG; protein mixed
.
protein disulfide bond; LOOH, lipid hydroperoxide; LOH,
. .
alcohol; R , secondary radical; DNA , DNA radical; M, metals; disulfide; Protein-SSX, protein disulfide bond; Protein-SH,
Protein-SH, protein sulhydril or protein thiol; PGG2, prostaglan- protein sulhydryl or protein thiol; Protein-SOH, Protein sulfenic
din G2; PGH2, prostaglandin H2; PES; Prostaglandin endoper- acid; Protein-SNO, protein nitrosothiol; Protein-S7, protein
.
oxide synthetase; GRX, glutaredoxin. thiolate anion; Protein S , protein thyil radical.
GSH in human health and disease 241

main non-enzymatic mechanisms for S-glutathiony- binding of GSH to endogenous compounds serves
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

lation of proteins involve: (1) reaction of GSH with to: (a) limit and regulate the activity of chemicals; (b)
partially oxidized protein-(SH) groups (sulfenic facilitate their membrane transport and elimination
acids); (2) protein-SH/GSSG exchange reactions; from the cell and organism; and (c) result in the
.
(3) protein-SH/GSH-GS radical reactions; and (4) formation of important intermediates (van Bladeren,
NO-mediated formation of S-nitrosoglutathione 2000; Townsend et al., 2005). Glutathione forms
.
(GSNO) and GS radicals (S-nitrosation or conjugates with a great variety of highly reactive
S-nitrosylation). It has been demonstrated that a electrophiles by either direct interaction (i.e. non-
significant basal level of protein mixed disulfides catalyzed); or by a reaction catalyzed by GST
(protein-SSG) of around 1% is found within cells, (S-glutathionylation). Glutathione reacts with var-
and where there is a significant increase in GSSG ious electrophiles (e.g. arene oxides, unsaturated
content due to oxidative stress, protein-SSG will carbonyls, organic halides), physiological metabolites
concomitantly increase. S-Glutathionylation has also (e.g. estrogen, melanins, prostaglandins and leuko-
been observed under basal conditions in the absence trienes), and xenobiotics (e.g. bromobenzene and
of oxidative insults. S-Glutathionylation of proteins acetaminophen). In this way, GSH forms thioether
(considered a reversible post-translational modifica- conjugates with compounds like leukotrienes, pros-
tion of proteins) has a dual role in protecting against taglandins, hepoxilins, nitric oxide hydroxyalkenals,
irreversible protein thiol oxidation and in regulating ascorbic acid, dopa, dopamine and maleic acid, while
protein function. Once formed, mixed disulfide it forms thioesthers with cysteine, coenzyme A,
linkages are removed by changes in the intracellular proteins and other cellular thiols (Eaton & Bammler,
GSH/GSSG balance and/or the activities of protein 1999; van Bladeren, 2000). Via non-catalyzed reac-
disulfide reductase, thioredoxin, glutaredoxin (thiol- tions GSH can also form thermodynamically stable
transferase) enzymes and other reducing agents but kinetically labile mercaptides with a high affinity
(dethiolation or deglutathionylation). The enzymatic for metals including mercury, silver, cadmium,
specificity and efficiency of dethiolating reactions has arsenic, lead, gold, zinc, and copper. In addition,
been observed to depend on the structural con- GSH also functions in the mobilization and delivery
formation of the disulfide (Pompella et al., 2003; of metals between ligands, in the transport of metal
Giustarini et al., 2004; Di Simplicio et al., 2005; across cell membranes, as a source of cysteine for
Shackelford et al., 2005; Biswas et al., 2006; Cross & metal binding, and as a reductant or cofactor in
Templeton, 2006). redox reactions involving metals (Wang & Ballatori,
1998).
The group of GSTs forms a class of enzymes with
Glutathionylation of electrophiles
overlapping substrate specificity, that generate a large
As a nucleophile, GSH can react with electrophiles set of thioesthers, called glutathione S-conjugates.
that are generated as a consequence of metabolic Most of the GST isoforms are present in the
processes involving both endogenous compounds cytoplasm, although some are also present in the
and xenobiotics (Figure 5, Table III). Conjugation of endoplasmic reticulum and mitochondrial matrix.
GSH is an essential aspect of both xenobiotic and This allows them to participate in the detoxification
normal physiological metabolism. In general, the of a chemically diverse group of compounds includ-
ing chemical carcinogens, environmental pollutants
and anti-tumor agents. The most common reactions
by GST involve nucleophilic attack by GSH to an
Table III. GSH-mediated conjugation reactions.
electrophilic carbon. These include those of satu-
Precursors Products rated (e.g. alkyl halides, lactones and epoxides) and
unsaturated carbon atoms (e.g. a, b-unsaturated
2GSH þ M2þ GS-M-SG compounds, quinones, quinonimines and esters) as
GSH þ PGA1 S-(PGA1)-GSH well as aromatic carbon atoms (e.g. aryl halides and
aryl nitro compounds) (van Bladeren, 2000). The
GSH þ Hepoxilin A3 Hepoxilin A3-C electrophilic centers of these compounds undergo
GSH þ 4-HNE 4-HNE-GSH nucleophilic substitution, nucleophilic addition to a
a, b-unsaturated ketones or epoxides, and nucleo-
GSH þ Dopamine þ 2O2 SS-gluathionyl-
philic attack on electrophilic oxygen (Townsend
dopamine þ 2HO2
GSH þ Methylgyoxal D-lactate þ GSH et al., 2005). Glutathione-S-transferase also mediates
formation of GSNO from alkyl nitrite and GSH.
2GSH þ 2ATP þ Trypanothione þ Moreover, GST can also serve as peroxidases,
Spermine 2ADP þ 2Pi isomerases and thiol transferases. In addition,
protein-protein interactions between GSTs and
Abbreviations: GSH, reduced glutathione; M, metal; PGA1,
prostaglandin A1; 4-HNE, 4-hydroxynonenal; GXI, glyoxylase I; signaling proteins such as JNK have been shown to
GXII, glyoxylase II; GSS; glutathionylspermidine synthetase; TS, regulate signal transduction (Pompella et al., 2003;
trypanothione synthetase. Hayes et al., 2005). Finally, GSH conjugation has
242 R. Franco et al.

been demonstrated to have a pro-oxidant activity efficiency of the DNA repair machinery (Smith et al.,
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

by itself that can give rise to the formation of 1996; Green, 2003; Green et al., 2006).
compounds that are at least as reactive as the parent
electrophiles. In this way, GSH-conjugates extrusion
Other biological functions of GSH
is an important mechanism of electrophile detoxifi-
cation although this may result in severe GSH Glutathione has been reported to participate in
depletion. In addition, GSH-conjugation of metal several other important biological processes. It also
ions has been reported to generate ROS including serves as a substrate for the formaldehyde dehydro-
.
O27 and H2O2 (Pompella et al., 2003). genase that converts formaldehyde and GSH to
S-formyl-glutathione. This is of physiological im-
portance because formaldehyde is a potent carcino-
Glutathione compartmentalization
gen produced from the metabolism of methionine,
Glutathione compartmentalization is also an impor- choline, methanol sarcosine, and xenobiotics.
tant process which participates in many other Glutathione is required as well for the conversion
biological processes. Significant pools are found of the arachidonic acid metabolite, prostaglandin H2,
within mitochondria, endoplasmic reticulum and into prostaglandins D2 and E2 by endoperoxide
nuclear matrix. Mitochondria are highly dependent isomerase. Furthermore, GSH is involved in the
on GSH for the prevention of oxidative damage due glyoxylase system, which converts methylgyoxal to
to aerobic respiration and they rely on the salvage of D-lactate in microorganisms. Finally, GSH is a
GSSG by GR and on the uptake of cytosolic GSH cofactor for transhydrogenases in the reduction of
across the outer membrane since they are incapable oxidized centers on DNA, proteins, and other
of de novo synthesis. It has been proposed that GSH biomolecules (Table III) (Meister, 1995b; Sies,
must be actively transported into the mitochondria or 1999; Jefferies et al., 2003; Wu et al., 2004).
exchanged for another anion not only because the
matrix space of the mitochondria possesses a negative
Glutathione as a central player in human
membrane potential relative to the cytoplasm, but
diseases
also because GSH is a negatively charged molecule at
physiological pH. Some of the possible candidates Alterations in GSH homeostasis and metabolism
involved in mitochondrial import of cytosolic GSH have been widely reported to be correlated with
include the monocarboxylate, dicarboxylate, 2-ox- several human diseases. Most of them have been
oglutarate, tricarboxylate, and glutamate-hydroxide associated with depletion of cellular GSH levels.
and glutamate-aspartate transporters (Figure 4) Glutathione depletion can be induced by three
(Lash, 2006). different mechanisms: (1) its oxidation by RS, (2)
In contrast to the cytosol and mitochondria, the its conjugation to proteins or electrophiles, and (3)
lumen of the endoplasmic reticulum (ER) contains a its extrusion across the plasma membrane. Under
relatively higher concentration of GSSG which these conditions, the cell demands GSH replenish-
allows the formation of disulfide bonds and their ment through de novo GSH synthesis or salvage
isomerization through the activity of protein disulfide pathways (catabolism). For example, GSH depletion
isomerases and oxidoreductases. The high concen- has been associated with the progression of cellular
tration of GSSG in the ER is maintained by the influx death and cytotoxicity. On the other hand, high
of reducing equivalents by either protein transloca- intracellular GSH concentrations are responsible for
tion, which provides with available cysteine residues, multidrug resistance observed in transformed (carci-
and/or selective transport of GSH from the cytosol to nogenous) cells. Finally, alterations in GSH-depen-
the ER. Protein disulfide isomerase (PDI) catalyzes dent enzymes have been described in different
the formation of disulfide bonds, then, the resulted disease conditions. In the next section, we briefly
reduced PDI is further oxidized by the flavoprotein summarize the role of alterations in intracellular
Ero1. The later generates ROS as byproducts that are GSH content and/or GSH-dependent enzymes or
detoxified by GSH thereby generating high levels of processes in the progression of several pathological
GSSG. Protein disulfide isomerase can be either conditions.
reduced by GSH or oxidized by GSSG. High luminal
GSSG concentrations are then secreted from the ER
Apoptosis
(Chakravarthi et al., 2006).
Nuclear matrix is another important pool for Apoptosis or programmed cell death is a ubiquitous,
GSH. No active transport mechanisms for GSH evolutionary, highly conserved process involved in
have been described so far for its nuclear import; several biological phenomena. Under physiological
however, it is assumed that GSH is capable of conditions, it is important not only in the constant
diffusing into the nucleus across nuclear pores. turnover of cells in all tissues but also during the
Recent reports have suggested the critical role of normal development and senescence of the organism.
nuclear GSH pools in protecting DNA from oxida- Moreover, its deregulation has been observed to
tive modifications that might, in turn, affect the occur as either a cause or consequence of distinct
GSH in human health and disease 243

pathologies (Fadeel & Orrenius, 2005). Apoptosis is a changes involved in the generation of a permissive
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

highly organized process characterized by the pro- apoptotic environment necessary for the activation
gressive activation of precise pathways leading to of apoptotic enzymes. Glutathione is essential for
specific biochemical and morphological alterations. survival as g-GCS knock-out mouse, which induces
Initial stages of apoptosis are characterized by RS GSH depletion, die from apoptotic cell death
formation, changes in intracellular ionic homeostasis, (Dalton et al., 2004). Several studies have shown a
cell shrinkage, loss of membrane lipid asymmetry and correlation between GSH depletion and the progres-
chromatin condensation. Later stages associated with sion of apoptosis (Figure 6), for example, inhibition
the execution phase of apoptosis are characterized by of GSH release is able to rescue cells from apoptosis
activation of execution caspases and endonucleases, (Ghibelli et al., 1998; Franco & Cidlowski, 2006;
apoptotic body formation and cell fragmentation Hammond et al., 2007). Moreover high [GSH]i
(Hengartner, 2000; Franco et al., 2006). levels have been associated with an apoptotic
Despite the large efforts on the characterization of resistant phenotype (Chiba et al., 1996; Kohno
the molecular events leading to apoptosis, recent et al., 1996; Mehlen et al., 1996; Watson et al.,
reports have underlined the role of changes in the 1997; Meredith et al., 1998; Xu et al., 1999; Friesen
intracellular milieu of the cells that are necessary for et al., 2004; Cazanave et al., 2006). Glutathione
the progression of the cell death program. Intracel- depletion during apoptosis has been associated with
lular GSH loss is an early hallmark in the progression the activation of a plasma membrane transport
of cell death in response to different apoptotic stimuli mechanism during apoptosis that is induced by both
(Beaver & Waring, 1995; Ghibelli et al., 1998; extrinsic and intrinsic apoptotic pathway activators
Obrosova et al., 1999; Hammond et al., 2004; Kern & (Ghibelli et al., 1995; van den Dobbelsteen et al.,
Kehrer, 2005), and it is thought to be part of the 1996; Bojes et al., 1997; Ghibelli et al., 1998;

Figure 6. Role of GSH in apoptosis and cytotoxicity. Glutathione depletion is an important hallmark for apoptosis and cytotoxicity, and can be
depleted by different mechanisms. Apoptosis induced by physiological stimuli such as death receptor activation or cytokine withdrawal has
been shown to induce GSH loss by the activation of a plasma membrane efflux transport for GSH and not by its oxidation to GSSG (1).
Distinct apoptotic stimuli (death receptors), stress conditions (osmotic, hypoxia), and cytotoxic agents (such as xenobiotics and pollutants)
can generate or increase reactive species (RS) formation either by organelle-mediated damage, or by simple chemical reactions, which can
induce GSH depletion through its oxidation (GSSG formation) and/or conjugation (2). GSH depletion and RS formation can trigger an
amplification cascade which utilizes both increased RS formation and GSH depletion (3). Mitochondrial GSH pool has also been shown to
act as an important regulator of cellular death. The resulted GSSG and/or GSH-conjugates generated are toxic to the cell and must be
extruded by efflux pumps. This further depletes intracellular GSH pools by the impairment of GSSG recycling to GSH through the action of
the GR and NADPH. Finally, GSH depletion and RS formation might regulate the induction of apoptosis or cytotoxicity by RS-mediated
signaling (through ROS or RNS), thiol-exchange reactions or protein oxidation-modifications (S-glutathionylation, S-nitrosylation), or GSH
conjugation (by the action of GST) (4). GSH-T, GSH transporter involved in GSH depletion during apoptosis.
244 R. Franco et al.

He et al., 2003; Hammond et al., 2004; Franco & a role of GSH content in the regulation of apoptotic
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

Cidlowski, 2006; Hammond et al., 2007). On the enzymes, rather than the involvement of RS.
other hand, oxidation of GSH by RS has been Glutathione depletion has been shown to either
demonstrated to mediate its depletion during predispose cells to apoptosis or directly trigger cell
apoptosis induced by cytotoxic agents that by them- death by modulation of both the permeability
selves induce oxidative stress (see next section) transition pore formation, and the activation of
(Hengartner, 2000; Fu et al., 2001; Hiroi et al., execution caspases (Ueda et al., 1998; Obrosova
2005; Cai et al., 2007). Recent studies have suggested et al., 1999; Coppola & Ghibelli, 2000; Haouzi et al.,
that both MRP (He et al., 2003; Benlloch et al., 2005; 2001; Armstrong & Jones, 2002; Nagai et al., 2002;
Hammond et al., 2007) and GSH/OA7 exchange Varghese et al., 2003; Valverde et al., 2006). In vitro
transporters might be involved in GSH efflux during studies have shown that a reduction in the GSH
apoptosis (Franco & Cidlowski, 2006). Additionally, content is necessary for the formation of the
a recent study has shown that glutamate-L-cysteine apoptosome (Sato et al., 2004). Additionally, GSH
ligase also known as g-glutamyl cysteine synthetase, depletion activates the intrinsic apoptotic pathway
the rate limiting enzyme in GSH biosynthesis, is a initiator Bax, by its oxidation-dependent dimeriza-
direct target of caspase 3 (Franklin et al., 2002). This tion (D’Alessio et al., 2005). It has also been
may represent another mechanism for diminishing proposed recently that released cytochrome C from
GSH levels during apoptosis. the mitochondria needs to become oxidized for its
The mechanisms involved in GSH-induced mod- pro-apoptotic activation, which will require cytosolic
ulation of apoptosis remain elusive. Due to its action GSH levels to be depleted (Coppola & Ghibelli,
as one of the main antioxidant defenses within the 2000; Hancock et al., 2003).
cell, depletion of intracellular GSH content has Several GSH-dependent enzymes have been re-
primarily been thought to reflect the generation of ported to protect cells from undergoing apoptosis.
RS (Wu et al., 2004). The role of RS in apoptosis has For example, overexpression of GPX has been
been extensively studied (Chandra et al., 2000; reported to protect against Fas-induced apoptosis
Carmody & Cotter, 2001). However, such role has (Gouaze et al., 2001). However, in vivo studies have
been suggested mainly through the use of RS shown that there are no difference in animals
generation systems that induce cell death. On the deficient of GPX compared to WT in their response
other hand, the role of RS in apoptosis induced by to death receptor-(Fas and TNF) mediated cell death
physiological stimuli, like that of death receptor (Bajt et al., 2002). Thiol exchange reactions have also
activation, remains largely controversial primarily been reported to directly modulate apoptotic en-
due to reports showing contradictory roles including zymes (England & Cotter, 2005). For example,
both inhibitory and stimulatory effects of RS on caspases have been reported to be glutathionylated
apoptosis. For example, apoptosis induced by and nitrosylated under basal conditions (Zech et al.,
activation of the death receptor Fas/CD95/Apo-1 by 1999; Klatt et al., 2000). Activation and/or transloca-
its ligand (FasL) has been suggested to depend on tion of these enzymes during apoptosis has been
the generation of H2O2 formation (Devadas et al., reported to require deglutathionylation (Pan & Berk,
2003; Perez-Cruz et al., 2003; Sato et al., 2004; 2007) and denitrosylation (Dimmeler et al., 1997;
Reinehr et al., 2005), while other studies report an Haendeler et al., 1997; Kim et al., 1997; Li et al.,
inhibitory role of H2O2 on Fas-mediated cell death 1997; Melino et al., 1997; Mohr et al., 1997; Kim
(Borutaite & Brown, 2001; Aronis et al., 2003). et al., 2000; Melino et al., 2000; Bernassola et al.,
Many reports have correlated ROS and Fas-induced 2001; Mannick et al., 2001; Kim & Tannenbaum,
apoptosis by the protective effect of extracellular 2004). Additionally, anti-apoptotic functions of Bcl-
thiols such as GSH and N-acetyl-L-cysteine (NAC) 2, FLICE inhibitory protein (FLIP) and dynamin
in preventing cell death, effects ascribed to their also depend on their degree of nitrosylation (Chan-
known capacities as antioxidants (Gulbins et al., vorachote et al., 2005; Moungjaroen et al., 2006;
1996; Um et al., 1996; Deas et al., 1997; Devadas Kang-Decker et al., 2007). Interestingly, inhibition of
et al., 2003; Sato et al., 2004; Franco & Cidlowski, GRX which regulates protein glutathionylation by
2006). Interestingly, protective effects of thiol-com- cadmium triggers apoptosis in the absence of any
pounds in the absence of any ROS formation are also other apoptotic stimuli (Chrestensen et al., 2000).
observed (Deas et al., 1997). Additionally, apoptosis
has been reported to occur under anaerobic
Stress response and cytotoxicity
conditions i.e. in the absence of ROS formation
(Jacobson & Raff, 1995; Shimizu et al., 1995), or in The intracellular content of GSH is highly responsive
cells lacking mitochondrial DNA, i.e. deficient in to environmental factors. Many different conditions
respiration and mitochondrial oxidative phosphory- of environmental stress have been demonstrated to
lation (Jacobson et al., 1993; Jiang et al., 1999). alter GSH content (Figure 6). These include heavy
Thus, inhibition of apoptosis by using any metals, high glucose concentrations, heat shock, RS
experimental intervention that prevents GSH or compounds that can generate RS. Modulation of
depletion or replenishes it might be an indicative of GSH content by xenobiotics has been shown to be
GSH in human health and disease 245

regulated by modulation of the g-GCS subunit gene that results in the production of free radicals and
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

ROS (GS ,  O27, H2O2) and oxidative stress


. .
expression by EpRE (electrophile response element,
known as well as antioxidant response element, (Paolicchi et al., 2002). Glutathione reductase
ARE), TRE (activatior protein-1) and TRE-like activity has also been suggested to play an important
transcription enhancer elements, which are modu- role in the enzymatic reduction of heavy metals
lated by oxidants, phenolic antioxidants, growth including Cr (VI) and vanadate (Ning & Grant,
factors, inflammatory and anti-inflammatory agents 2000; Gunaratnam & Grant, 2001). In contrast,
in various cells (Dickinson & Forman, 2002). GSH supplementation (via NAC, GSH or GSH-
Exposure to agents such as 4-hydroxy-2-nonenal monoester), alone or in combination with other
(4-HNE) increase the content of GSH by increasing antioxidants, has been shown to have protective
the rate of GSH synthesis, which facilitates 4-HNE effects against experimental models of iron-induced
detoxification from GST. The GSH-conjugate or neurodegeneration, chromium picolate-induced
adduct of 4-HNE is by itself an inhibitor of GST, and toxicity, cobalt particle-induced inflammation, ar-
has been suggested to be detoxified from the cell by senic-induced lipid peroxidation, and nickel-induced
RLIP76. 4-Hydroxy-2-nonenal has also been de- oxidative stress (Valko et al., 2005; Valko et al.,
monstrated to activate the JNK pathways, which in 2006). Cell death mediated by oxidative stress from
turn activate the activator protein-1 (AP-1) transcrip- arsenic exposure has been demonstrated to be largely
tion complex that can further regulate GSH bio- prevented by GSH or NAC (Choi et al., 2002; Hu
synthesis (Sharma et al., 2004; Yadav et al., 2007). et al., 2003; Kang et al., 2003; McCafferty-Grad
Oxidant-mediated damage has been widely re- et al., 2003; Baysan et al., 2007; Habib et al., 2007;
ported as the main mechanism of metal-induced Piga et al., 2007; Santra et al., 2007). In addition,
cytotoxicity. Iron (Fe)-, copper (Cu)-, chromium overexpression of GST has also been shown to be
(Cr)-, cobalt (Co)-, vanadium (Va)-, cadmium protective by decreasing arsenic trioxide-induced
(Cd)-, arsenic (As)- and nickel (Ni)-mediated H2O2 formation (Lu et al., 2004; Zhou et al., 2005;
toxicity have been widely reported induced by free Habib et al., 2007). Finally, GSH supplementation of
radicals-mediated DNA damage, lipid peroxidation cells with NAC prevents apoptosis induced by heavy
and changes in calcium and sulphydril homeostasis metals including cadmium (Oh & Lim, 2006),
(Valko et al., 2005; Valko et al., 2006). Arsenic- chromium (Hayashi et al., 2004) and copper (Zhai
induced GSH depletion is one of the most studied et al., 2000).
models for metal-induced toxicity. Studies so far Cellular stress has also been widely reported to
have suggested three distinct mechanisms by which induce apoptosis primarily through oxidant toxicity.
arsenic depletes cellular GSH pools: (1) via GSH- In general, GSH depletion has been shown to
mediated reduction of pentavalent to trivalent sensitize cells to compounds that produce oxidative
arsenicals where GSH acts as an electron donor; cytolysis (Meister, 1991). Overexpression of
(2) via direct interaction of GSH with arsenic; and PHGPX and GSTs has been reported to be
(3) by its oxidation through arsenic-induced genera- protective against apoptosis induced by ultraviolet
tion of free radicals (Kumagai & Sumi, 2007). On the radiation (UV), cytokine withdrawal, stress and
other hand, both beneficial and harmful effects of xenobiotics which is associated with lipid peroxida-
GSH metabolism have been reported during metal- tion and RS formation (Nomura et al., 2001;
induced toxicity. For example, Chromium (VI) does Sharma et al., 2004; Briz et al., 2006). Additionally,
not react with DNA in vitro or in isolated nuclei, until overexpression of the g-GCS protects HEK293 cells
it reacts with cellular reductants, causing a wide against UV irradiation-induced cell death by inhibi-
variety of DNA lesions including Cr – DNA adducts, tion of the JNK1 and caspase-3 activation (Fan
DNA – protein crosslinks, DNA – DNA crosslinks et al., 2005). Accordingly, GSH supplementation
and oxidative damage. Glutathione has been shown with NAC also prevents apoptosis induced by UV
to rapidly form a complex with Cr(VI), followed by a radiation (Bush et al., 1999), chemotherapeutics
slow reduction of Cr(VI) to yield Cr(V) leading to (Park et al., 2000; Custodio et al., 2002; Kurosu
.
the formation of GS . In addition to the cellular et al., 2003; Jiang et al., 2007), drugs (Rosati et al.,
. .
damaging effect of the GS radical, GS can react 2004) and cytokine withdrawal (Kirkland &
further with other thiol molecules in oxygenated Franklin, 2001).
conditions to give O27. Furthermore, Cr(V) can be
.

.
reduced by GSH to Cr(IV) generating OH . It has
Cancer
also been demonstrated that in the presence of Co
(II), Fe(III), Ni(II) and Cu(II), endogenous reduc- Numerous factors contribute to the initiation, devel-
tants such as GSH cause DNA damage by exacer- opment and progression of cancer including those of
bation of oxidative stress (Valko et al., 2005; genetic, environmental and dietary influences.
Valko et al., 2006). Similarly, the reduction of Fe(III) Glutathione and GSH-metabolism participate in
to Fe(II) by GSH in particular has been suggested different ways in cancer prevention and progression
to be mediated through the g-GT-mediated GSH (Figure 7). Both clinical and epidemiological studies
cleavage that triggers an iron-redox cycling process have shown the role of RS produced by UV radiation
246 R. Franco et al.
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

Figure 7. Role of GSH in carcinogenesis. Glutathione has been shown to regulate the progression of carcinogenesis in different ways. Its
protective role in cellular transformation involves the detoxification of carcinogens (RS or xenobiotics) produced by UV exposure, chemical
agents, environment, inflammation and diet. This is mediated either by its antioxidant role or by GST-mediated phase II detoxification
reactions (1). Transformed cells have shown to have increased levels of expression for several GSH enzymes whose function could be also
altered by polymorphisms. Glutathione inhibition of apoptosis and thiol/exchange reactions also contribute to cellular transformation and
immortalization. In addition, in tumoral cells, abnormal levels of several ROS and RNS regulate post-translational modifications through
S-glutathionylation or S-nitrosylation of redox-sensitive transcription factors and proteins that have been shown to be involved in
tumorigenesis (2). Finally, GSH also contributes to cell’s resistance to antineoplastic treatments by either inhibition of apoptosis, or
impairment of chemotherapeutic drug detoxification by GSH-conjugation (3). In this way, therapeutic strategies based on GSH depletion
(by its extrusion or metabolism) have been shown to sensitize cells to anti-cancer therapy.

exposure, chemical carcinogens, environmental modifications through S-glutathionylation and/or


agents, inflammation and diet, in cancer etiology. S-nitrosylation of redox-sensitive transcription fac-
By its antioxidant properties, GSH participates in the tors (e.g. NF-kB, Oxy R and AP-1) and enzymes
defense against carcinogens by protecting cells from (e.g. PKC and Ras) that have been reported to be
ROS-mediated DNA-damage and ROS-induced involved in tumorigenesis (England & Cotter, 2005;
regulation of gene expression (Valko et al., 2006). Biswas et al., 2006; Fukumura et al., 2006).
Reduced GR and GPX activity has been reported in Glutathione metabolism has also been reported to
tumor tissues (Tanriverdi et al., 2007). In addition, participate in chemotherapy resistance. Increased
GPX polymorphisms have been associated with expression of GSTs and GSH-transporters as well
cancer (Moscow et al., 1994). Glutathione can also as high GSH concentration are common features of
protects cells from potential carcinogens by GST- transformed cells that, in turn, are associated with
mediated phase II detoxification reactions that high resistance to chemotherapeutic agents (Yang
mediate the conjugation of GSH with potential et al., 2006b). When antineoplastic or chemother-
carcinogenic compounds (Rosado et al., 2006). apeutic drugs enter cancer cells, they are conjugated
Recent studies have as well linked GST polymorph- with glutathione and are excreted through GSH-
isms that are associated with reduced enzymatic pumps of the MRP family of transporters. It is known
activity and increased risk of blood, bladder, gastric, that MRP transporters are highly expressed in
colorectal, skin, breast, kidney and liver cancers malignant cells. Thus, inhibitors of MRP-mediated
(McIlwain et al., 2006). Moreover, redox regulation transport of GSH-conjugates have been largely used
of thiol groups has also been involved in carcino- as a common adjuvant in anti-cancer therapy (Cole &
genesis. For instance, many malignant cell types Deeley, 2006). High intracellular GSH content in
posses abnormal levels of several ROS and RNS tumor cells is a consequence of high g-GCS expres-
(Giles, 2006) that when combined with GSH result sion and can promote tumor cell survival by inhibi-
in increases in GSSG and GSNO concentrations. tion of apoptotic cell death activation. In accordance,
This might further mediate post-translational modulation of intracellular GSH content has been
GSH in human health and disease 247

largely studied for potential anti-cancer therapies. mediated by oxidative stress has been shown to play
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

Such therapeutic interventions have been shown to an important role in regulating transient outward Kþ
increase cellular sensitivity to treatment with ionizing currents in rat ventricular myocytes that might
radiation and also decrease tumor resistance to mediate impaired contractility and abnormal elec-
chemotherapeutic agents (Balendiran et al., 2004). trical properties during diabetes (Xu et al., 2002).
Furthermore, GSH depletion sensitizes tumor cells to Despite that GSH depletion has been suggested to be
undergo apoptosis overriding Bcl-2 protection and a cause of free radicals formation, such depletion has
thus suggesting a link between GSH content and the also been associated to its metabolism independent
anti-apoptotic function of the Bcl-2 family of anti- from its oxidation (Beard et al., 2003; Januel et al.,
apoptotic proteins (Kern & Kehrer, 2005). Another 2003).
potential role for anti-neoplastic therapy is the The mitochondrial GSH pool also seems to be an
modulation of GSH efflux by acting on its transpor- important player in diabetes. Hyperglycemia has
ters or pumps. Recently, it has been shown that been shown to increase the enzymatic conversion of
transformed cells are sensitized to cell death when glucose to sorbitol by the sorbitol dehydrogenase,
intracellular GSH is depleted through stimulation of with concomitant decreases in NADPH and glu-
GSH efflux pumps (MRP, CFTR and OATP tathione. This depletion in reducing equivalents
proteins) (Estrela et al., 2006; Franco & Cidlowski, results in augmented sensitivity to oxidative stress
2006), whose expression varies in different cancer cell (Brownlee, 2003; Rolo & Palmeira, 2006). Diabetic
lines (Cui et al., 2003; Perez-Tomas, 2006). streptozotocin rats have been reported to have
reduced GPX activity, and increased levels of GSSG
and ROS in the mitochondria (Awasthi et al., 2003;
Diabetes
Green et al., 2004; Ghosh et al., 2005; Mastrocola
Metabolic diseases arise either when a critical et al., 2005). Hyperglycemia-induced oxidative stress
enzyme is disabled, or if a control mechanism for a and GSH depletion have been suggested to be
metabolic pathway is affected. Diabetes mellitus is a regulated by the inner membrane potential of the
metabolic disease characterized by hyperglycemia mitochondria (Gustafsson et al., 2004), and oxidative
and oxidative stress (Maritim et al., 2003; Green stress has been reported to impair insulin secretion
et al., 2004; Opara, 2004; Pennathur & Heinecke, (Krippeit-Drews et al., 1999; Maechler et al., 1999).
2004; Robertson, 2004; Robertson et al., 2004). Nicotinamide nucleotide transhydrogenase (NNT)
Enhanced production of free radicals and oxidative has been demonstrated to protect against mitochon-
stress play a central role in the pathogenesis of drial oxidative stress by mediating NADPþ and
diabetes and its complications, including obesity, NADH conversion to NADPH that is necessary for
which is the most critical factor in the emergence of GSSG recycling to GSH (Arkblad et al., 2005).
metabolic diseases. However, controversy still exists Recently, it has been proposed that alterations in
about the nature, magnitude, and localization of NNT activity in pancreatic b cells mediate impaired
oxidative stress in diabetes. Moreover, it is still insulin secretion during type 2 diabetes. The
unclear whether oxidative stress is a primary event proposed mechanism involves the impairment of
that occurs early in the progression of diabetes or if it GR-mediated GSH recycling after its oxidation that
is a secondary phenomenon as part of the end-stage is required for ROS-detoxification by GPX. This is
tissue damage (Pennathur & Heinecke, 2004). thought to lead to enhanced ROS accumulation
Several studies have demonstrated that diabetes decreased ATP production and enhanced KATP
elicits dynamic and profound alterations in GSH channel activity (ATP-inhibited Kþ channels) there-
metabolism and transport, that mediate alterations in by producing hyperpolarization of the plasma mem-
the cell’s redox state (Figure 8) (Gandhi & Roy brane and preventing Ca2þ-dependent insulin
Chowdhury, 1979; Murakami et al., 1989; Collier secretion (Freeman et al., 2006a; Freeman et al.,
et al., 1990; Paolisso et al., 1992; Yoshida et al., 1995; 2006b).
Lu et al., 1997; Giron et al., 1999; Obrosova et al., Although GST polymorphisms together with
2000; Sharma et al., 2000; Yang et al., 2000; Abou- altered GST distribution and activity have been
Seif & Youssef, 2001; Yue et al., 2003; Raza & John, reported to occur in distinct models of diabetes, their
2004; Winiarska et al., 2004; Darmaun et al., 2005; physiological significance is still elusive (Fujita et al.,
Sampathkumar et al., 2005). Several pathological 2001; Traverso et al., 2002; Raza et al., 2004; Wang
complications of diabetes including peripheral nerve et al., 2006; Yalin et al., 2006). In this respect, insulin
lesions, embryonic malformations, angiopathies- and treatment has been shown to up-regulate the anti-
artherosclerosis-mediated tissue hypoxia, diabetic oxidant defenses of the cells by increasing GST
encephalopathy, and gastric damage have been expression through the PI3K/Akt/p70S6K pathway
proposed to be associated with GSH depletion (Kim et al., 2003; Kim et al., 2006), while glucagon
(Goldin et al., 1997; Sakamaki et al., 1999; Gumie- on the other hand, a physiological antagonist of
niczek et al., 2001; Donma et al., 2002; Mastrocola insulin, decreases GST expression (Kim et al., 2003).
et al., 2005; Bonfigli et al., 2006; Ho et al., 2006; Additionally, insulin treatment in patients with type 2
Mehta et al., 2006). For example, GSH depletion diabetes mellitus reduces intracellular oxidative
248 R. Franco et al.
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

Figure 8. Mechanisms for ROS formation and GSH depletion during hyperglycemia. During diabetes, elevated plasma glucose and free fatty acids
(FFA) lead to a rise in its cytoplasmic concentration by its uptake and diffusion respectively. Increase glucose and FFA oxidation lead to
increased production of reducing equivalents that mediate ROS generation through the respiratory electron transport chain (1). This is
followed by a concomitant decrease in mitochondrial NADPH and GSH. NNT mutation or deficiency has been proposed as a possible
mechanism involved in the impairment of NADPH formation and by consequence GR-mediated GSH recycling after its oxidation, which is
necessary for ROS-detoxification by GPX (2). This leads to increased ROS accumulation that have been shown to decrease ATP and
enhance KATP channels producing hyperpolarization of the plasma membrane and preventing Ca2þ-dependent insulin secretion (3).
Reactive oxygen species have been proposed to act as the major mechanism mediating cytosolic GSH depletion, although other mechanisms
including GSH efflux might also contribute to its depletion (4). Oxidative stress and GSH depletion might further alter insulin signaling
through thiol/exchange reactions. Diabetes has also been shown to decrease the expression of GSH-dependent antioxidant enzymes such as
GST and GPX (5). Finally, GSH depletion might also contribute to FFA and ROS-mediated apoptosis in b-cells (6). GLUT4, glucose
transporter; ETC, electron transport chain; NNT, Nicotinamide nucleotide transhydrogenase; ATP-S, ATP synthase.

stress through an increased GSH/GSSG ratio (Bravi sensitization (McClung et al., 2004). Thiol-exchange
et al., 2006), while GSH infusion in type 2 diabetes reactions might also play an important role in
mellitus increases insulin sensitivity (De Mattia et al., diabetes. Cellular levels of protein S-nitrosylation
1998). Glutathione reductase activity has also been and S-glutathionylation have been demonstrated to
reported to be protective against streptozotocin- be elevated in type 2 diabetic patients (Niwa et al.,
induced diabetes (Nagaoka et al., 2004). In addition, 2000; Sampathkumar et al., 2005; Kaneki et al.,
diabetes has been reported to either impair GPX 2007). Additionally, S-nitrosylation of the insulin
activity (Bhor et al., 2004; Manea et al., 2004; Sindhu receptor, insulin receptor substrate-1, and Akt/PKB,
et al., 2004), which will decrease the antioxidant have been reported in skeletal muscle of diabetic
defenses; or to increase GPX activity as a compensa- mice where the Akt/PKB pathway was shown to be
tory response to ROS formation (Ulusu et al., 2003). reversibly inactivated (Yasukawa et al., 2005).
Glutathione peroxidase activity has been observed to A reduction in the number of insulin producing
have both beneficial and deleterious effects on b-cells through apoptosis also contributes to the
diabetes. For example GPX overexpression has been progression of diabetes. Hyperglycemia has been
shown to protect pancreatic beta cells from oxidative shown to induce apoptosis via activation of NFkB,
stress induced by glucose toxicity (Tanaka et al., mitochondrial cytochrome C, formation of ROS,
2002; Robertson et al., 2003). However, a recent and overexpression of the apoptotic genes Bad, Bik
report demonstrates a development of insulin resis- and Bid (Federici et al., 2001; Mandrup-Poulsen,
tance in mammals with elevated expression of the 2003). Additionally, free fatty acid-induced b-cell
antioxidant enzyme GPX1 whose activity may inter- apoptosis is suggested to involve the formation of
fere with insulin function by over quenching in- ceramide and NO. Finally, high glucose has been
tracellular ROS that are required for insulin shown to mediate upregulation of Fas in human
GSH in human health and disease 249

islets which might induce apoptosis in neighboring Glutathione deficiency has been largely correlated
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

b-cells constitutively expressing FasL (Mandrup- with pulmonary diseases including chronic pulmon-
Poulsen, 2003). Altered GSH homeostasis and ary disease, acute respiratory distress syndrome,
oxidative stress in diabetes are also important neonatal lung damage and asthma. Glutathione
factors modulating cell death in different tissues. levels in the epithelial lining fluid (ELF), the first
As explained above, GSH is a central regulator of line of defense against exposure to oxygen, airborne
apoptosis. MRP-mediated GSH efflux and mito- toxins and oxygen radicals released from lung
chondrial GSH depletion are required for caspase 9- phagocytes, were observed to be depleted in idio-
and 3-dependent cardiomyocyte apoptosis in short pathic pulmonary fibrosis and acute distress syn-
term diabetic rats (Ghosh et al., 2004; Ghosh et al., drome. Moreover, GSH depletion in the ELF
2005). Furthermore, NAC supplementation pro- contributes to the imbalance between oxidants and
tects these cells from oxidative stress-mediated antioxidants in the lungs amplifying inflammatory
injury (Fiordaliso et al., 2004; Patriarca et al., responses and potentiating lung damage. It is
2005). Finally, it has been demonstrated that skin possible that GSH differences in ELF in various
cells derived from obese mouse are more sensitive inflammatory lung diseases are due to changes in the
to oxidative stress induced by UV radiation molecular regulation of GSH synthesis and the
(Katiyar & Meeran, 2007). production of lipid peroxidation products (Rahman
et al., 2005). Glutathione content in lung cells has
also been shown to be critical to the lung antioxidant
Glutathione synthetase deficiency
defenses against oxidant/free radical (cigarette
Glutathione synthetase deficiency is a rare autosomal smoke/air particulate)-mediated injury and inflam-
recessive disorder which prevents the production of mation. Air pollution gases such as sulfur dioxide
GSH due to a mutation and malfunctioning of the and nitrogen dioxide generate an inflammatory
GS as the result of a mutation in the corresponding response and increase alveolar permeability by
gene (Njalsson & Norgren, 2005). Glutathione depleting GSH levels in the lung. Additionally, it
synthetase deficiency can be classified into three has been suggested that extracellular GPX secreted
types based on the severity of the disease. Mild GS into ELF by alveolar epithelial cells and macro-
deficiency results in the destruction of red blood cells phages, along with the GSH redox system, provide a
and concomitant hemolytic anemia. These patients strong defense against oxidants (Rahman, 2005).
also excrete large amounts of 5-oxoproline in their It is known that supplemental oxygen can cause
urine (5-oxoprolinuria), which builds up when GSH significant lung damage in prematurely born infants
is not processed correctly in cells (Al-Jishi et al., which puts them at risk for developing pulmonary
1999), while individuals with moderate GSH defi- oxygen toxicity. This is mainly ascribed to cellular
ciency also present elevated acidity in the blood damage which is probably mediated by increased
and tissues (metabolic acidosis). Patients suffering production of ROS in the mitochondria. Protection
from the severe form of this disorder experience against these ROS is provided mainly by GSH, and
recurrent bacterial infections, and neurological depletion of mitochondrial GSH has been asso-
symptoms including seizures, generalized slowing ciated with an increased susceptibility to oxidant
down of physical reactions, movements, and injury (O’Donovan & Fernandes, 2000). Cystic
speech (psychomotor retardation), mental retarda- fibrosis is a genetic disorder ascribed to a recessive
tion, and a loss of coordination (ataxia) (Ristoff & mutation of the organic anion efflux channel
Larsson, 1998; Njalsson, 2005). More than 20 CFTR. CFTR contributes to the cellular homeo-
mutations in the GS gene have been described in static balance of GSH and in this way cystic
patients suffering from GS deficiency. These muta- fibrosis is associated with systemic and cellular
tions change single amino acids in the GS or disrupt GSH deficiency, by altered GSH efflux. GSH
how genetic information from the GS gene is deficiency in these conditions has been observed
spliced to make a blueprint for producing this to regulate inflammatory cytokines through NFkB
enzyme. The mutated GS enzyme is either unstable, activation and to promote high levels of oxidative
shorter than usual, or in the wrong conformation, stress and lipid peroxidation byproducts (Hudson,
and in all cases shows reduced activity that disrupts 2001).
the g-glutamyl cycle and production of GSH
(Njalsson et al., 2005).
Other human diseases and pathological conditions
Alterations in GSH homeostasis have also been
Lung diseases
described in several other pathological conditions;
Lungs are amongst the major sources of GSH however, its implication in the pathogenesis of these
storage (6.1 – 17.5 nmol/mg lung) and have higher conditions is still unclear. For example, GSH has
levels of g-GCS than other tissues. Alterations in the been found to be consistently lower in children with
levels of reduced GSH in the lung lining fluid have oedematous protein energy malnutrition (PEM)
been shown in various inflammatory conditions. (such as in kwashiorkor and marasmic-kwashiorkor)
250 R. Franco et al.

compared with non-oedematous PEM, suggesting a The aging process has also been associated with
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

direct relation with oedematous malnutrition. It has deterioration of GSH homeostasis by increased
been suggested that the limited cysteine content in levels of GSSG with a concomitant reduction of
the diet is the underlying cause for the reduced rate intracellular GSH. Additionally, disruption of the
of GSH synthesis in these patients (Reid & Jahoor, microsomal GST gene significantly reduces life-
2001). span in Drosophila suggesting a role of lipid
Glutathione levels are also depleted in plasma, peroxidation in cell aging and senescence. A down-
epithelial lining fluid, peripheral blood mononuclear regulation of the g-GCS has been reported to occur
lymphocytes and monocytes of symptom-free HIV in the rat brain during aging. In the brain, a
seropositive individuals, asymptomatic HIV-infected complex interaction between astrocytes and neurons
individuals and AIDS patients. However, the me- exists, where astrocytes supply neurons with cy-
chanisms by which alterations in GSH metabolism steine for GSH synthesis that is necessary for the
regulate HIV progression remain unclear. Glu- defense against oxidative-stress based pathologies
tathione deficiency has been associated with HIV including those of Parkinson’s, Alzheimer’s and
infection. HIV-viral maturation depends on protease Huntington’s disease, as well as in ischemia,
activity regulated by glutathionylation. Additionally, schizophrenia and epilepsy. Parkinson’s disease
depleted GSH levels are suggested to activate NFkB progression is associated with a depletion of GSH
which allows HIV expression. Finally, GSH deple- levels and an increase in ROS formation in the
tion has also been suggested to be a modulator of substantia nigra. Finally, some reports have also
apoptosis in CD4þ T-cell lymphocytes which pre- attributed a signaling role to GSH as a hormone
sent low GSH content during HIV progression. In (Dringen & Hirrlinger, 2003).
this way, NAC-based treatment for replenishing
GSH stores has been demonstrated to have beneficial
Modulation of GSH content as a means of
effects (Droge & Breitkreutz, 2000; Shackelford
therapeutic approach
et al., 2005).
Liver is one of the major pools of GSH, and Because of the relevance of GSH as a central element
alterations in its content are either the cause or in the regulation of several human diseases, there is
consequence of several pathologies. Alcoholism- an increasing interest in the design of new strategies
mediated depletion of mitochondrial GSH mediates to modulate the GSH content. As indicated earlier,
ROS-dependent cell death (triggered by cytokines), several pathological conditions are associated with
that in turn contribute to the progression of GSH depletion either by its increased metabolism,
cirrhosis. This has been reported to be associated transport or impaired synthesis. In this way ther-
to the inactivation of the mitochondrial import apeutic treatments have been focused on the
transport for GSH (Fernandez-Checa & Kaplowitz, supplementation of cells and tissues with GSH.
2005). Additionally, chronic hepatitis C infection is Dietary amino acid balance has an important effect
associated with depleted levels of GSH in hepatic on GSH homeostasis. In particular, the adequate
and plasma fractions, as well as in peripheral blood provision of sulfur-containing amino acids as well as
mononuclear cells. Acetaminophen in addition to glutamate (glutamine) and glycine (serine) is critical
other pharmaceutical and/or environmental xeno- for optimal GSH synthesis (Valencia et al., 2001a;
biotics can deplete liver GSH. Finally, GSH levels Valencia et al., 2001e; Valencia et al., 2001c; Parcell,
after partial hepatectomy have been reported to be 2002; Wu et al., 2004). Glutamine is an efficient
doubled due to increased g-GCS expression and precursor for glutamate involved in GSH synthesis.
activity. In this conditions, it has been proposed that Cysteine is not only the rate limiting precursor to
an increased reducing environment caused by GSH but it is by itself an important reducing agent.
increased GSH may be generated in order to ensure Cysteine can be given orally to increase GSH;
proper NFkB activity and hepatocyte survival however, cysteine has been shown to have toxic
(Han et al., 2006). effects due to its instability and pro-oxidant reac-
Like GS deficiency, g-GCS deficiency is also tions. Because cysteine can be generated via the
associated with haemolytic anemia. On the other trans-sulfuration pathway, dietary methionine
hand g-GT deficiency exhibits marked glutathionuria (through the synthesis of S-adenosylmethionine)
and glutathionaemia (excess glutathione in the can replace cysteine to support GSH synthesis
blood), which can lead to an imbalance in glutamic in vivo (Wu et al., 2004). Administration of oral
acid homeostasis (Schulman et al., 1975). Patients GSH increases hepatic GSH levels; however, the
with Crohn’s disease and ulcerative colitis present mechanisms involved in its absorption are still
low level activities of g-GCT, g-GT and GST unclear. Glutathione is thought to be taken up poorly
(Jefferies et al., 2003). On the other hand, perturba- by cells and rapidly degraded in the gut and
tions in S-glutathionylated protein homeostasis is circulation. Moreover, GSH does not readily cross
associated with hyperlipidemia, chronic renal failure, the blood – brain barrier. In this way, intravenous or
uremia, as well as with Friederich’s ataxia and oral administration of GSH serves only to provide
Down’s syndrome (Giustarini et al., 2004). brain cells with constituent amino acids. Indeed,
GSH in human health and disease 251

GSH has to be broken down into its amino acids,


Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

Conclusions and perspectives


transported into the cell and then resynthesized.
Thus, the use of GSH precursors is a more efficient Glutathione is a central component of many meta-
way to increase GSH levels (Valencia & Hardy, 2002; bolic pathways. Studies so far have uncovered that
Njalsson & Norgren, 2005). GSH is not only one of the most important
Precursors of GSH include NAC, GSH antioxidant defenses and a major regulator of the
monoethyl or diethyl esters, and 2-oxothiazolidine- redox balance in the cell, but it is also involved in a
4-carboxylate (OTC or procysteine). N-acetyl- myriad of other cellular processes. It is a key
cysteine is a derivative of the sulfur-containing mediator of many physiological events including
amino acid cysteine and an intermediary (along cellular signaling, detoxification, and thiol disulfide
with glutamic acid and glycine) in the conversion of exchange reactions. Moreover, its deregulation has
cysteine to GSH. It is intracellularly hydrolyzed to been associated with a wide range of pathological
cysteine before it serves as a precursor for GSH conditions such as AIDS, ischemia, neurodegenera-
formation and has limited toxicity compared to tive, metabolic, and inherited diseases. Throughout
other thiols. Oral NAC administration leads to an this review article, we have highlighted the role of
increase in intracellular cysteine and GSH levels, GSH in the pathophysiology of several human
however, intravenous administration is preferably diseases where glutathione metabolism has shown
recommended since nausea and vomiting limit its to play a transcendental role in disease progression.
effectiveness in oral therapy. Glutathione ethyl Clearly, it becomes apparent the importance in
esters are lipid soluble compounds that are readily designing pharmacologically-based strategies aimed
transported into cells where they are de-esterified for specific targets in the metabolism, transport and
by esterases and increase the levels of GSH. catabolism of GSH, as potential therapeutic treat-
Glutathione diethyl ester is more rapidly trans- ments against human diseases.
ported into cells than the monoethyl ester. 2-
Oxothiazolidine-4-carboxylate is transported into
Acknowledgements
the cell and metabolized forming cysteine; however,
OTC and other thiazolidines are of limited value This work was supported, in part, by the Intramural
because of the by-products formed such as for- Research Program of the NIH/National Institute of
maldehyde. It is important to mention that pre- Environmental Health Sciences (Franco and Scho-
cursors of cysteine or its dipeptides have been neveld); by the European Community through a
shown to have limited usefulness in raising GSH Marie Curie International Reintegration Grant with-
levels in patients with GS deficiency or altered acti- in the 6th European Community Framework Pro-
vity (Wernerman & Hammarqvist, 1999; Schafer & gram (Pappa); and the American Institute for Cancer
Buettner, 2001; Valencia et al., 2002). Research (AICR) through a Marilyn Gentry Fellow-
High intracellular GSH levels are also important ship (Panayiotidis).
contributors to pathologies such as cellular transfor-
mation and resistance to antineoplastic treatments in
cancer cells. In this way, other tools are available References
in order to decrease intracellular GSH levels. Abou-Seif MA, Youssef AA. 2001. Oxidative stress and male
L-buthionine-SR-sulfoximide (BSO) has been widely IGF-1, gonadotropin and related hormones in diabetic
used as a specific inhibitor of g-GCS thereby patients. Clin Chem Lab Med 39:618 – 23.
inhibiting the synthesis of g-glutamylcysteine and Al-Jishi E, Meyer BF, Rashed MS, Al-Essa M, Al-Hamed MH,
Sakati N, Sanjad S, Ozand PT, Kambouris M. 1999.
the formation of GSH. This experimental approach Clinical, biochemical, and molecular characterization of
has been shown to efficiently deplete cells from patients with glutathione synthetase deficiency. Clin Genet 55:
cytosolic GSH, although mitochondrial GSH has 444 – 9.
been observed to remain largely intact. Thus, a Arkblad EL, Tuck S, Pestov NB, Dmitriev RI, Kostina MB,
Stenvall J, Tranberg M, Rydstrom J. 2005. A Caenorhabditis
combination of BSO and thiol oxidants such as
elegans mutant lacking functional nicotinamide nucleotide
diethylmalate (DEM) or diamide, have proven transhydrogenase displays increased sensitivity to oxidative
efficient in depleting cells from intracellular GSH, stress. Free Radic Biol Med 38:1518 – 25.
although coupled to a high degree of cytotoxicity. Armstrong JS, Jones DP. 2002. Glutathione depletion enforces
Ethacrynic acid is another compound reported to the mitochondrial permeability transition and causes cell
deplete cells from GSH via GST. It has been death in Bcl-2 overexpressing HL60 cells. Faseb J 16:
1263 – 5.
reported to deplete both cytosolic and mitochon- Aronis A, Melendez JA, Golan O, Shilo S, Dicter N, Tirosh O.
drial GSH. Moreover, 1, 3-Bis(2-chloroethyl)-N- 2003. Potentiation of Fas-mediated apoptosis by attenuated
nitrosourea (BCNU, or carmustine) is another production of mitochondria-derived reactive oxygen species.
compound that carbonylates the lysine residue in Cell Death Differ 10:335 – 44.
the active site of GR. This NADPH-dependent Awasthi S, Singhal SS, Sharma R, Zimniak P, Awasthi YC. 2003.
Transport of glutathione conjugates and chemotherapeutic
reaction is fast and inhibits GR, thereby preventing drugs by RLIP76 (RALBP1): a novel link between G-protein
the recycling of GSSG to GSH (Schafer & Buettner, and tyrosine kinase signaling and drug resistance. Int J Cancer
2001). 106:635 – 46.
252 R. Franco et al.

Bajt ML, Ho YS, Vonderfecht SL, Jaeschke H. 2002. Reactive Cazanave S, Berson A, Haouzi D, Vadrot N, Fau D, Grodet A,
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

oxygen as modulator of TNF and fas receptor-mediated Letteron P, Feldmann G, El-Benna J, Fromenty B, et al. 2007.
apoptosis in vivo: studies with glutathione peroxidase-deficient High hepatic glutathione stores alleviate Fas-induced apoptosis
mice. Antioxid Redox Signal 4:733 – 40. in mice. J Hepatol 46:858 – 68.
Balendiran GK, Dabur R, Fraser D. 2004. The role of glutathione Chakravarthi S, Jessop CE, Bulleid NJ. 2006. The role of
in cancer. Cell Biochem Funct 22:343 – 52. glutathione in disulphide bond formation and endoplasmic-
Ballatori N, Hammond CL, Cunningham JB, Krance SM, reticulum-generated oxidative stress. EMBO Rep 7:271 – 5.
Marchan R. 2005. Molecular mechanisms of reduced glu- Chandra J, Samali A, Orrenius S. 2000. Triggering and modulation
tathione transport: role of the MRP/CFTR/ABCC and OATP/ of apoptosis by oxidative stress. Free Radic Biol Med 29:
SLC21A families of membrane proteins. Toxicol Appl 323 – 33.
Pharmacol 204:238 – 55. Chanvorachote P, Nimmannit U, Wang L, Stehlik C, Lu B,
Baysan A, Yel L, Gollapudi S, Su H, Gupta S. 2007. Arsenic Azad N, Rojanasakul Y. 2005. Nitric oxide negatively regulates
trioxide induces apoptosis via the mitochondrial pathway by Fas CD95-induced apoptosis through inhibition of ubiquitin-
upregulating the expression of Bax and Bim in human B cells. proteasome-mediated degradation of FLICE inhibitory pro-
Int J Oncol 30:313 – 18. tein. J Biol Chem 280:42044 – 50.
Beard KM, Shangari N, Wu B, O’Brien PJ. 2003. Metabolism, Chiba T, Takahashi S, Sato N, Ishii S, Kikuchi K. 1996. Fas-
not autoxidation, plays a role in alpha-oxoaldehyde- and mediated apoptosis is modulated by intracellular glutathione in
reducing sugar-induced erythrocyte GSH depletion: human T cells. Eur J Immunol 26:1164 – 9.
relevance for diabetes mellitus. Mol Cell Biochem 252: Choi YJ, Park JW, Suh SI, Mun KC, Bae JH, Song DK, Kim SP,
331 – 8. Kwon TK. 2002. Arsenic trioxide-induced apoptosis in U937
Beaver JP, Waring P. 1995. A decrease in intracellular glutathione cells involve generation of reactive oxygen species and
concentration precedes the onset of apoptosis in murine inhibition of Akt. Int J Oncol 21:603 – 10.
thymocytes. Eur J Cell Biol 68:47 – 54. Chrestensen CA, Starke DW, Mieyal JJ. 2000. Acute cadmium
Benlloch M, Ortega A, Ferrer P, Segarra R, Obrador E, exposure inactivates thioltransferase (Glutaredoxin), inhibits
Asensi M, Carretero J, Estrela JM. 2005. Acceleration intracellular reduction of protein-glutathionyl-mixed disulfides,
of glutathione efflux and inhibition of gamma- and initiates apoptosis. J Biol Chem 275:26556 – 65.
glutamyltranspeptidase sensitize metastatic B16 melanoma Cole SP, Deeley RG. 2006. Transport of glutathione and gluta-
cells to endothelium-induced cytotoxicity. J Biol Chem thione conjugates by MRP1. Trends Pharmacol Sci 27:438 – 46.
280:6950 – 59. Collier A, Wilson R, Bradley H, Thomson JA, Small M. 1990.
Bernassola F, Catani MV, Corazzari M, Rossi A, Melino G. 2001. Free radical activity in type 2 diabetes. Diabet Med 7:27 – 30.
Inactivation of multiple targets by nitric oxide in CD95- Contreras JE, Sanchez HA, Veliz LP, Bukauskas FF, Bennett MV,
triggered apoptosis. J Cell Biochem 82:123 – 33. Saez JC. 2004. Role of connexin-based gap junction channels
Bhor VM, Raghuram N, Sivakami S. 2004. Oxidative damage and and hemichannels in ischemia-induced cell death in nervous
altered antioxidant enzyme activities in the small intestine of tissue. Brain Res Brain Res Rev 47:290 – 303.
streptozotocin-induced diabetic rats. Int J Biochem Cell Biol Coppola S, Ghibelli L. 2000. GSH extrusion and and the
36:89 – 97. mitochondrial pathway of apoptotic signalling. Biochem Soc
Biswas S, Chida AS, Rahman I. 2006. Redox modifications of Trans 28:56 – 61.
protein-thiols: emerging roles in cell signaling. Biochem Cross JV, Templeton DJ. 2006. Regulation of signal transduction
Pharmacol 71:551 – 64. through protein cysteine oxidation. Antioxid Redox Signal
Bojes HK, Datta K, Xu J, Chin A, Simonian P, Nunez G, 8:1819 – 27.
Kehrer JP. 1997. Bcl-xL overexpression attenuates glutathione Cui Y, Konig J, Nies AT, Pfannschmidt M, Hergt M, Franke WW,
depletion in FL5.12 cells following interleukin-3 withdrawal. Alt W, Moll R, Keppler D. 2003. Detection of the human organic
Biochem J 325(Part 2):315 – 19. anion transporters SLC21A6 (OATP2) and SLC21A8
Bonfigli A, Colafarina S, Falone S, Di Giulio C, Di Ilio C, (OATP8) in liver and hepatocellular carcinoma. Lab Invest
Amicarelli F. 2006. High levels of antioxidant enzymatic 83:527 – 38.
defence assure good protection against hypoxic stress in Custodio JB, Cardoso CM, Almeida LM. 2002. Thiol protecting
spontaneously diabetic rats. Int J Biochem Cell Biol 38: agents and antioxidants inhibit the mitochondrial permeability
2196 – 208. transition promoted by etoposide: implications in the preven-
Borutaite V, Brown GC. 2001. Caspases are reversibly inactivated tion of etoposide-induced apoptosis. Chem Biol Interact
by hydrogen peroxide. FEBS Lett 500:114 – 18. 140:169 – 84.
Bravi MC, Armiento A, Laurenti O, Cassone-Faldetta M, D’Alessio M, De Nicola M, Coppola S, Gualandi G, Pugliese L,
De Luca O, Moretti A, De Mattia G. 2006. Insulin decreases Cerella C, Cristofanon S, Civitareale P, Ciriolo MR,
intracellular oxidative stress in patients with type 2 diabetes Bergamaschi A, et al. 2005. Oxidative Bax dimerization
mellitus. Metabolism 55:691 – 5. promotes its translocation to mitochondria independently of
Briz O, Romero MR, Martinez-Becerra P, Macias RI, Perez MJ, apoptosis. Faseb J 19:1504 – 6.
Jimenez F, San Martin FG, Marin JJ. 2006. OATP8/1B3- Dalton TP, Chen Y, Schneider SN, Nebert DW, Shertzer HG.
mediated cotransport of bile acids and glutathione: an export 2004. Genetically altered mice to evaluate glutathione
pathway for organic anions from hepatocytes? J Biol Chem homeostasis in health and disease. Free Radic Biol Med
281:30326 – 35. 37:1511 – 26.
Brownlee M. 2003. A radical explanation for glucose-induced beta Darmaun D, Smith SD, Sweeten S, Sager BK, Welch S,
cell dysfunction. J Clin Invest 112:1788 – 90. Mauras N. 2005. Evidence for accelerated rates of glutathione
Bush JA, Ho VC, Mitchell DL, Tron VA, Li G. 1999. Effect of utilization and glutathione depletion in adolescents with poorly
N-acetylcysteine on UVB-induced apoptosis and DNA repair in controlled type 1 diabetes. Diabetes 54:190 – 6.
human and mouse keratinocytes. Photochem Photobiol 70: De Mattia G, Bravi MC, Laurenti O, Cassone-Faldetta M,
329 – 33. Armiento A, Ferri C, Balsano F. 1998. Influence of reduced
Cai X, Pan N, Zou G. 2007. Copper-1, 10-phenanthroline- glutathione infusion on glucose metabolism in patients with non-
induced apoptosis in liver carcinoma bel-7402 cells associates insulin-dependent diabetes mellitus. Metabolism 47:993 – 7.
with copper overload, reactive oxygen species production, Deas O, Dumont C, Mollereau B, Metivier D, Pasquier C,
glutathione depletion and oxidative DNA damage. Biometals Bernard-Pomier G, Hirsch F, Charpentier B, Senik A. 1997.
20:1 – 11. Thiol-mediated inhibition of FAS and CD2 apoptotic
Carmody RJ, Cotter TG. 2001. Signalling apoptosis: a radical signaling in activated human peripheral T cells. Int Immunol
approach. Redox Rep 6:77 – 90. 9:117 – 25.
GSH in human health and disease 253

Devadas S, Hinshaw JA, Zaritskaya L, Williams MS. 2003. Fas- Friesen C, Kiess Y, Debatin KM. 2004. A critical role of
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

stimulated generation of reactive oxygen species or exogenous glutathione in determining apoptosis sensitivity and resis-
oxidative stress sensitize cells to Fas-mediated apoptosis. Free tance in leukemia cells. Cell Death Differ 11 Suppl 1:
Radic Biol Med 35:648 – 61. S73 – 85.
Di Simplicio P, Frosali S, Priora R, Summa D, Cherubini Di Fu Y, Sies H, Lei XG. 2001. Opposite roles of selenium-
Simplicio F, Di Giuseppe D, Di Stefano A. 2005. Biochemical dependent glutathione peroxidase-1 in superoxide generator
and biological aspects of protein thiolation in cells and plasma. diquat- and peroxynitrite-induced apoptosis and signaling.
Antioxid Redox Signal 7:951 – 63. J Biol Chem 276:43004 – 9.
Dickinson DA, Forman HJ. 2002. Cellular glutathione and thiols Fujita H, Haseyama T, Kayo T, Nozaki J, Wada Y, Ito S,
metabolism. Biochem Pharmacol 64:1019 – 26. Koizumi A. 2001. Increased expression of glutathione
Dimmeler S, Haendeler J, Nehls M, Zeiher AM. 1997. Suppres- S-transferase in renal proximal tubules in the early stages of
sion of apoptosis by nitric oxide via inhibition of interleukin- diabetes: a study of type-2 diabetes in the Akita mouse model.
1beta-converting enzyme (ICE)-like and cysteine protease Exp Nephrol 9:380 – 6.
protein (CPP)-32-like proteases. J Exp Med 185:601 – 7. Fukumura D, Kashiwagi S, Jain RK. 2006. The role of nitric
Donma O, Yorulmaz E, Pekel H, Suyugul N. 2002. Blood and oxide in tumour progression. Nat Rev Cancer 6:521 –
lens lipid peroxidation and antioxidant status in normal 34.
individuals, senile and diabetic cataractous patients. Curr Eye Gandhi CR, Roy Chowdhury D. 1979. Effect of diabetes mellitus
Res 25:9 – 16. on sialic acid & glutathione content of human erythrocytes of
Dringen R, Hirrlinger J. 2003. Glutathione pathways in the brain. different ages. Indian J Exp Biol 17:585 – 7.
Biol Chem 384:505 – 16. Garcia-Ruiz C, Fernandez-Checa JC, Kaplowitz N. 1992. Bidir-
Droge W, Breitkreutz R. 2000. Glutathione and immune function. ectional mechanism of plasma membrane transport of reduced
Proc Nutr Soc 59:595 – 600. glutathione in intact rat hepatocytes and membrane vesicles.
Eaton DL, Bammler TK. 1999. Concise review of the glutathione J Biol Chem 267:22256 – 64.
S-transferases and their significance to toxicology. Toxicol Sci Ghibelli L, Coppola S, Rotilio G, Lafavia E, Maresca V,
49:156 – 64. Ciriolo MR. 1995. Non-oxidative loss of glutathione in
England K, Cotter TG. 2005. Direct oxidative modifications of apoptosis via GSH extrusion. Biochem Biophys Res Commun
signalling proteins in mammalian cells and their effects on 216:313 – 20.
apoptosis. Redox Rep 10:237 – 45. Ghibelli L, Fanelli C, Rotilio G, Lafavia E, Coppola S, Colussi C,
Estrela JM, Ortega A, Obrador E. 2006. Glutathione in cancer Civitareale P, Ciriolo MR. 1998. Rescue of cells from
biology and therapy. Crit Rev Clin Lab Sci 43:143 – 81. apoptosis by inhibition of active GSH extrusion. Faseb
Fadeel B, Orrenius S. 2005. Apoptosis: a basic biological J 12:479 – 86.
phenomenon with wide-ranging implications in human disease. Ghosh S, Pulinilkunnil T, Yuen G, Kewalramani G, An D, Qi D,
J Intern Med 258:479 – 517. Abrahani A, Rodrigues B. 2005. Cardiomyocyte apoptosis
Fan Y, Wu D, Jin L, Yin Z. 2005. Human glutamylcysteine induced by short-term diabetes requires mitochondrial
synthetase protects HEK293 cells against UV-induced cell GSH depletion. Am J Physiol Heart Circ Physiol 289:H768 –
death through inhibition of c-Jun NH2-terminal kinase. Cell 76.
Biol Int 29:695 – 702. Ghosh S, Ting S, Lau H, Pulinilkunnil T, An D, Qi D,
Federici M, Hribal M, Perego L, Ranalli M, Caradonna Z, Abrahani MA, Rodrigues B. 2004. Increased efflux of
Perego C, Usellini L, Nano R, Bonini P, Bertuzzi F, et al. 2001. glutathione conjugate in acutely diabetic cardiomyocytes. Can
High glucose causes apoptosis in cultured human pancreatic J Physiol Pharmacol 82:879 – 87.
islets of Langerhans: a potential role for regulation of specific Giles GI. 2006. The redox regulation of thiol dependent signaling
Bcl family genes toward an apoptotic cell death program. pathways in cancer. Curr Pharm Des 12:4427 – 43.
Diabetes 50:1290 – 301. Giron MD, Salto R, Gonzalez Y, Giron JA, Nieto N, Periago JL,
Fernandez-Checa JC, Kaplowitz N. 2005. Hepatic mitochondrial Suarez MD, Hortelano P. 1999. Modulation of hepatic and
glutathione: transport and role in disease and toxicity. Toxicol intestinal glutathione S-transferases and other antioxidant
Appl Pharmacol 204:263 – 73. enzymes by dietary lipids in streptozotocin diabetic rats.
Fiordaliso F, Bianchi R, Staszewsky L, Cuccovillo I, Doni M, Chemosphere 38:3003 – 13.
Laragione T, Salio M, Savino C, Melucci S, Santangelo F, Giustarini D, Rossi R, Milzani A, Colombo R, Dalle-Donne I.
et al. 2004. Antioxidant treatment attenuates hyperglycemia- 2004. S-glutathionylation: from redox regulation of
induced cardiomyocyte death in rats. J Mol Cell Cardiol protein functions to human diseases. J Cell Mol Med 8:
37:959 – 68. 201 – 12.
Forman HJ, Torres M, Fukuto J. 2002. Redox signaling. Mol Cell Goldin E, Ardite E, Elizalde JI, Odriozola A, Panes J, Pique JM,
Biochem 234 – 235:49 – 62. Fernandez-Checa JC. 1997. Gastric mucosal damage in
Franco R, Bortner CD, Cidlowski JA. 2006. Potential roles of experimental diabetes in rats: role of endogenous glutathione.
electrogenic ion transport and plasma membrane depolariza- Gastroenterology 112:855 – 63.
tion in apoptosis. J Membr Biol 209:43 – 58. Gouaze V, Mirault ME, Carpentier S, Salvayre R, Levade T,
Franco R, Cidlowski JA. 2006. SLCO/OATP-like transport of Andrieu-Abadie N. 2001. Glutathione peroxidase-1 overex-
glutathione in FasL-induced apoptosis: glutathione efflux is pression prevents ceramide production and partially inhibits
coupled to an organic anion exchange and is necessary for the apoptosis in doxorubicin-treated human breast carcinoma
progression of the execution phase of apoptosis. J Biol Chem cells. Mol Pharmacol 60:488 – 96.
281:29542 – 57. Green DR. 2003. Overview: apoptotic signaling pathways in the
Franklin CC, Krejsa CM, Pierce RH, White CC, Fausto N, immune system. Immunol Rev 193:5 – 9.
Kavanagh TJ. 2002. Caspase-3-Dependent Cleavage of the Green K, Brand MD, Murphy MP. 2004. Prevention of
Glutamate-L-Cysteine Ligase Catalytic Subunit during Apop- mitochondrial oxidative damage as a therapeutic strategy in
totic Cell Death. Am J Pathol 160:1887 – 94. diabetes. Diabetes 53 Suppl 1:S110 – 8.
Freeman H, Shimomura K, Cox RD, Ashcroft FM. 2006a. Green RM, Graham M, O’Donovan MR, Chipman JK,
Nicotinamide nucleotide transhydrogenase: a link between Hodges NJ. 2006. Subcellular compartmentalization of glu-
insulin secretion, glucose metabolism and oxidative stress. tathione: correlations with parameters of oxidative stress
Biochem Soc Trans 34:806 – 10. related to genotoxicity. Mutagenesis 21:383 – 90.
Freeman H, Shimomura K, Horner E, Cox RD, Ashcroft FM. Gukasyan HJ, Lee VH, Simityan H, Kim KJ, Kannan R. 2006.
2006b. Nicotinamide nucleotide transhydrogenase: a key role Thermodynamic stoichiometry of Naþ-coupled glutathione
in insulin secretion. Cell Metab 3:35 – 45. transport. Can J Physiol Pharmacol 84:1223 – 7.
254 R. Franco et al.

Gulbins E, Brenner B, Schlottmann K, Welsch J, Heinle H, Hu XM, Hirano T, Oka K. 2003. Arsenic trioxide induces
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

Koppenhoefer U, Linderkamp O, Coggeshall KM, Lang F. apoptosis in cells of MOLT-4 and its daunorubicin-resistant
1996. Fas-induced programmed cell death is mediated by a cell line via depletion of intracellular glutathione, disruption of
Ras-regulated O2- synthesis. Immunology 89:205 – 12. mitochondrial membrane potential and activation of caspase-3.
Gumieniczek A, Hopkala H, Wojtowicz Z, Nieradko M. 2001. Cancer Chemother Pharmacol 52:47 – 58.
Differences in antioxidant status in skeletal muscle tissue in Hudson VM. 2001. Rethinking cystic fibrosis pathology: the
experimental diabetes. Clin Chim Acta 314:39 – 45. critical role of abnormal reduced glutathione (GSH) trans-
Gunaratnam M, Grant MH. 2001. The role of glutathione port caused by CFTR mutation. Free Radic Biol Med
reductase in the cytotoxicity of chromium (VI) in isolated rat 30:1440 – 61.
hepatocytes. Chem Biol Interact 134:191 – 202. Iantomasi T, Favilli F, Marraccini P, Magaldi T, Bruni P,
Gustafsson H, Soderdahl T, Jonsson G, Bratteng JO, Forsby A. Vincenzini MT. 1997. Glutathione transport system in human
2004. Insulin-like growth factor type 1 prevents hyperglycemia- small intestine epithelial cells. Biochim Biophys Acta
induced uncoupling protein 3 down-regulation and oxidative 1330:274 – 83.
stress. J Neurosci Res 77:285 – 91. Jacobson MD, Burne JF, King MP, Miyashita T, Reed JC,
Gutierrez-Correa J, Stoppani AO. 1997. Inactivation of yeast Raff MC. 1993. Bcl-2 blocks apoptosis in cells lacking
glutathione reductase by Fenton systems: effect of metal mitochondrial DNA. Nature 361:365 – 9.
chelators, catecholamines and thiol compounds. Free Radic Jacobson MD, Raff MC. 1995. Programmed cell death and Bcl-2
Res 27:543 – 55. protection in very low oxygen. Nature 374:814 – 6.
Habib GM, Shi ZZ, Lieberman MW. 2007. Glutathione protects Januel C, Fay LB, Ruggiero D, Lagarde M, Vericel E. 2003.
cells against arsenite-induced toxicity. Free Radic Biol Med Covalent coupling of reduced glutathione with ribose: loss of
42:191 – 201. cosubstrate ability to glutathione peroxidase. Biochim Biophys
Haendeler J, Weiland U, Zeiher AM, Dimmeler S. 1997. Effects of Acta 1620:125 – 32.
redox-related congeners of NO on apoptosis and caspase-3 Jefferies H, Coster J, Khalil A, Bot J, McCauley RD, Hall JC.
activity. Nitric Oxide 1:282 – 93. 2003. Glutathione. ANZ J Surg 73:517 – 22.
Hagenbuch B, Meier PJ. 2004. Organic anion transporting Jiang M, Wei Q, Pabla N, Dong G, Wang CY, Yang T, Smith SB,
polypeptides of the OATP/SLC21 family: phylogenetic Dong Z. 2007. Effects of hydroxyl radical scavenging on
classification as OATP/SLCO superfamily, new nomenclature cisplatin-induced p53 activation, tubular cell apoptosis and
and molecular/functional properties. Pflugers Arch 447:653 – nephrotoxicity. Biochem Pharmacol 73:1499 – 510.
65. Jiang S, Cai J, Wallace DC, Jones DP. 1999. Cytochrome
Hammond CL, Lee TK, Ballatori N. 2001. Novel roles for c-mediated apoptosis in cells lacking mitochondrial DNA.
glutathione in gene expression, cell death, and membrane Signaling pathway involving release and caspase 3 activation is
transport of organic solutes. J Hepatol 34:946 – 54. conserved. J Biol Chem 274:29905 – 11.
Hammond CL, Madejczyk MS, Ballatori N. 2004. Activation of Jones DP. 2006. Redefining oxidative stress. Antioxid Redox
plasma membrane reduced glutathione transport in death Signal 8:1865 – 79.
receptor apoptosis of HepG2 cells. Toxicol Appl Pharmacol Kaneki M, Shimizu N, Yamada D, Chang K. 2007. Nitrosative
195:12 – 22. Stress and Pathogenesis of Insulin Resistance. Antioxid Redox
Hammond CL, Marchan R, Krance SM, Ballatori N. 2007. Glu- Signal 9:319 – 29.
tathione export during apoptosis requires functional multidrug Kang-Decker N, Cao S, Chatterjee S, Yao J, Egan LJ, Semela D,
resistance-associated proteins. J Biol Chem 282:14337 – 47. Mukhopadhyay D, Shah V. 2007. Nitric oxide promotes
Han D, Hanawa N, Saberi B, Kaplowitz N. 2006. Mechanisms of endothelial cell survival signaling through S-nitrosylation and
liver injury. III. Role of glutathione redox status in liver injury. activation of dynamin-2. J Cell Sci 120:492 – 501.
Am J Physiol Gastrointest Liver Physiol 291:G1 – 7. Kang SH, Song JH, Kang HK, Kang JH, Kim SJ, Kang HW,
Hancock JT, Desikan R, Neill SJ. 2003. Cytochrome c, Lee YK, Park DB. 2003. Arsenic trioxide-induced apoptosis is
glutathione, and the possible role of redox potentials in independent of stress-responsive signaling pathways but
apoptosis. Ann N Y Acad Sci 1010:446 – 8. sensitive to inhibition of inducible nitric oxide synthase in
Haouzi D, Lekehal M, Tinel M, Vadrot N, Caussanel L, HepG2 cells. Exp Mol Med 35:83 – 90.
Letteron P, Moreau A, Feldmann G, Fau D, Pessayre D. Katiyar SK, Meeran SM. 2007. Obesity increases the risk of UV
2001. Prolonged, but not acute, glutathione depletion pro- radiation-induced oxidative stress and activation of MAPK and
motes Fas-mediated mitochondrial permeability transition and NF-kappaB signaling. Free Radic Biol Med 42:299 – 310.
apoptosis in mice. Hepatology 33:1181 – 8. Kern JC, Kehrer JP. 2005. Free radicals and apoptosis: relation-
Hayashi Y, Kondo T, Zhao QL, Ogawa R, Cui ZG, Feril LB, Jr., ships with glutathione, thioredoxin, and the BCL family of
Teranishi H, Kasuya M. 2004. Signal transduction of p53- proteins. Front Biosci 10:1727 – 38.
independent apoptotic pathway induced by hexavalent chro- Kim JE, Tannenbaum SR. 2004. S-Nitrosation regulates the
mium in U937 cells. Toxicol Appl Pharmacol 197:96 – 106. activation of endogenous procaspase-9 in HT-29 human colon
Hayes JD, Flanagan JU, Jowsey IR. 2005. Glutathione trans- carcinoma cells. J Biol Chem 279:9758 – 64.
ferases. Annu Rev Pharmacol Toxicol 45:51 – 88. Kim SK, Abdelmegeed MA, Novak RF. 2006. Identification of the
He YY, Huang JL, Ramirez DC, Chignell CF. 2003. Role of insulin signaling cascade in the regulation of alpha-class
reduced glutathione efflux in apoptosis of immortalized human glutathione S-transferase expression in primary cultured rat
keratinocytes induced by UVA. J Biol Chem 278:8058 – 64. hepatocytes. J Pharmacol Exp Ther 316:1255 – 61.
Hengartner MO. 2000. The biochemistry of apoptosis. Nature Kim SK, Woodcroft KJ, Novak RF. 2003. Insulin and
407:770 – 6. glucagon regulation of glutathione S-transferase expression in
Hiroi M, Ogihara T, Hirano K, Hasegawa M, Morinobu T, primary cultured rat hepatocytes. J Pharmacol Exp Ther 305:
Tamai H, Niki E. 2005. Regulation of apoptosis by glutathione 353 – 61.
redox state in PC12 cells exposed simultaneously to iron and Kim YM, Kim TH, Chung HT, Talanian RV, Yin XM,
ascorbic acid. Free Radic Biol Med 38:1057 – 72. Billiar TR. 2000. Nitric oxide prevents tumor necrosis factor
Ho EC, Lam KS, Chen YS, Yip JC, Arvindakshan M, alpha-induced rat hepatocyte apoptosis by the interruption of
Yamagishi S, Yagihashi S, Oates PJ, Ellery CA, Chung SS, mitochondrial apoptotic signaling through S-nitrosylation of
Chung SK. 2006. Aldose reductase-deficient mice are pro- caspase-8. Hepatology 32:770 – 8.
tected from delayed motor nerve conduction velocity, in- Kim YM, Talanian RV, Billiar TR. 1997. Nitric oxide
creased c-Jun NH2-terminal kinase activation, depletion of inhibits apoptosis by preventing increases in caspase-3-like
reduced glutathione, increased superoxide accumulation, and activity via two distinct mechanisms. J Biol Chem 272:
DNA damage. Diabetes 55:1946 – 53. 31138 – 48.
GSH in human health and disease 255

Kirkland RA, Franklin JL. 2001. Evidence for redox regulation of Maritim AC, Sanders RA, Watkins JB, 3rd. 2003. Diabetes,
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

cytochrome C release during programmed neuronal death: oxidative stress, and antioxidants: a review. J Biochem Mol
antioxidant effects of protein synthesis and caspase inhibition. Toxicol 17:24 – 38.
J Neurosci 21:1949 – 63. Masella R, Di Benedetto R, Vari R, Filesi C, Giovannini C.
Klatt P, Pineda Molina E, Perez-Sala D, Lamas S. 2000. Novel 2005. Novel mechanisms of natural antioxidant compounds
application of S-nitrosoglutathione-Sepharose to identify pro- in biological systems: involvement of glutathione and
teins that are potential targets for S-nitrosoglutathione-induced glutathione-related enzymes. J Nutr Biochem 16:577 – 86.
mixed-disulphide formation. Biochem J 349:567 – 78. Mastrocola R, Restivo F, Vercellinatto I, Danni O, Brignardello E,
Kohno T, Yamada Y, Hata T, Mori H, Yamamura M, Aragno M, Boccuzzi G. 2005. Oxidative and nitrosative stress in
Tomonaga M, Urata Y, Goto S, Kondo T. 1996. Relation brain mitochondria of diabetic rats. J Endocrinol 187:37 – 44.
of oxidative stress and glutathione synthesis to CD95(Fas/ Mates JM. 2000. Effects of antioxidant enzymes in the molecular
APO-1)-mediated apoptosis of adult T cell leukemia cells. control of reactive oxygen species toxicology. Toxicology
J Immunol 156:4722 – 8. 153:83 – 104.
Krippeit-Drews P, Kramer C, Welker S, Lang F, Ammon HP, McCafferty-Grad J, Bahlis NJ, Krett N, Aguilar TM, Reis I,
Drews G. 1999. Interference of H2O2 with stimulus-secretion Lee KP, Boise LH. 2003. Arsenic trioxide uses caspase-
coupling in mouse pancreatic beta-cells. J Physiol 514 (Part dependent and caspase-independent death pathways in myelo-
2):471 – 81. ma cells. Mol Cancer Ther 2:1155 – 64.
Kumagai Y, Sumi D. 2007. Arsenic: signal transduction, McClung JP, Roneker CA, Mu W, Lisk DJ, Langlais P, Liu F,
transcription factor, and biotransformation involved in cellular Lei XG. 2004. Development of insulin resistance and obesity in
response and toxicity. Annu Rev Pharmacol Toxicol 47:243 – mice overexpressing cellular glutathione peroxidase. Proc Natl
62. Acad Sci USA 101:8852 – 7.
Kurosu T, Fukuda T, Miki T, Miura O. 2003. BCL6 over- McIlwain CC, Townsend DM, Tew KD. 2006. Glutathione
expression prevents increase in reactive oxygen species and S-transferase polymorphisms: cancer incidence and therapy.
inhibits apoptosis induced by chemotherapeutic reagents in Oncogene 25:1639 – 48.
B-cell lymphoma cells. Oncogene 22:4459 – 68. Mehlen P, Kretz-Remy C, Preville X, Arrigo AP. 1996. Human
Lash LH. 2005. Role of glutathione transport processes in kidney hsp27, Drosophila hsp27 and human alphaB-crystallin
function. Toxicol Appl Pharmacol 204:329 – 42. expression-mediated increase in glutathione is essential for
Lash LH. 2006. Mitochondrial glutathione transport: physiologi- the protective activity of these proteins against TNFalpha-
cal, pathological and toxicological implications. Chem Biol induced cell death. Embo J 15:2695 – 706.
Interact 163:54 – 67. Mehta JL, Rasouli N, Sinha AK, Molavi B. 2006. Oxidative stress in
Lee TK, Hammond CL, Ballatori N. 2001. Intracellular glu- diabetes: a mechanistic overview of its effects on atherogenesis
tathione regulates taurocholate transport in HepG2 cells. and myocardial dysfunction. Int J Biochem Cell Biol 38:794 –
Toxicol Appl Pharmacol 174:207 – 15. 803.
Li J, Billiar TR, Talanian RV, Kim YM. 1997. Nitric oxide Meister A. 1991. Glutathione deficiency produced by inhibition of
reversibly inhibits seven members of the caspase family via its synthesis, and its reversal; applications in research and
S-nitrosylation. Biochem Biophys Res Commun 240: therapy. Pharmacol Ther 51:155 – 94.
419 – 24. Meister A. 1994. Glutathione, ascorbate, and cellular protection.
Li L, Lee TK, Meier PJ, Ballatori N. 1998. Identification of Cancer Res 54:1969s – 1975s.
glutathione as a driving force and leukotriene C4 as a substrate Meister A. 1995a. Glutathione biosynthesis and its inhibition.
for oatp1, the hepatic sinusoidal organic solute transporter. Methods Enzymol 252:26 – 30.
J Biol Chem 273:16184 – 91. Meister A. 1995b. Glutathione metabolism. Methods Enzymol
Li L, Meier PJ, Ballatori N. 2000. Oatp2 mediates bidirectional 251:3 – 7.
organic solute transport: a role for intracellular glutathione. Melino G, Bernassola F, Knight RA, Corasaniti MT, Nistico G,
Mol Pharmacol 58:335 – 40. Finazzi-Agro A. 1997. S-nitrosylation regulates apoptosis.
Lu M, Xia L, Luo D, Waxman S, Jing Y. 2004. Dual effects of Nature 388:432 – 3.
glutathione-S-transferase pi on As2O3 action in prostate cancer Melino G, Catani MV, Corazzari M, Guerrieri P, Bernassola F.
cells: enhancement of growth inhibition and inhibition of 2000. Nitric oxide can inhibit apoptosis or switch it into
apoptosis. Oncogene 23:3945 – 52. necrosis. Cell Mol Life Sci 57:612 – 22.
Lu SC, Kuhlenkamp J, Wu H, Sun WM, Stone L, Kaplowitz N. Meredith MJ, Cusick CL, Soltaninassab S, Sekhar KS,
1997. Progressive defect in biliary GSH secretion in Lu S, Freeman ML. 1998. Expression of Bcl-2 increases
streptozotocin-induced diabetic rats. Am J Physiol 272: intracellular glutathione by inhibiting methionine-dependent
G374 – 82. GSH efflux. Biochem Biophys Res Commun 248:458 – 63.
Maechler P, Jornot L, Wollheim CB. 1999. Hydrogen peroxide Mohr S, Zech B, Lapetina EG, Brune B. 1997. Inhibition of
alters mitochondrial activation and insulin secretion in caspase-3 by S-nitrosation and oxidation caused by nitric
pancreatic beta cells. J Biol Chem 274:27905 – 13. oxide. Biochem Biophys Res Commun 238:387 – 91.
Maellaro E, Del Bello B, Sugherini L, Comporti M, Casini AF. Moscow JA, Schmidt L, Ingram DT, Gnarra J, Johnson B,
1997. Purification and characterization of glutathione-depen- Cowan KH. 1994. Loss of heterozygosity of the human cytosolic
dent dehydroascorbate reductase from rat liver. Methods glutathione peroxidase I gene in lung cancer. Carcinogenesis
Enzymol 279:30 – 5. 15:2769 – 73.
Mahagita C, Grassl SM, Piyachaturawat P, Ballatori N. 2007. Moungjaroen J, Nimmannit U, Callery PS, Wang L, Azad N,
Human organic anion transporter 1B1 (OATP1B1/OATP-C) Lipipun V, Chanvorachote P, Rojanasakul Y. 2006. Reactive
and 1B3 (OATP1B3/OATP-8) function as bidirectional oxygen species mediate caspase activation and apoptosis induced
carriers and do not mediate GSH-bile acid co-transport. Am by lipoic acid in human lung epithelial cancer cells through Bcl-2
J Physiol Gastrointest Liver Physiol 293:G271 – 8. down-regulation. J Pharmacol Exp Ther 319:1062 – 9.
Mandrup-Poulsen T. 2003. Apoptotic signal transduction path- Murakami K, Kondo T, Ohtsuka Y, Fujiwara Y, Shimada M,
ways in diabetes. Biochem Pharmacol 66:1433 – 40. Kawakami Y. 1989. Impairment of glutathione metabolism in
Manea A, Constantinescu E, Popov D, Raicu M. 2004. Changes erythrocytes from patients with diabetes mellitus. Metabolism
in oxidative balance in rat pericytes exposed to diabetic 38:753 – 8.
conditions. J Cell Mol Med 8:117 – 26. Nagai H, Matsumaru K, Feng G, Kaplowitz N. 2002. Reduced
Mannick JB, Schonhoff C, Papeta N, Ghafourifar P, Szibor M, glutathione depletion causes necrosis and sensitization to
Fang K, Gaston B. 2001. S-Nitrosylation of mitochondrial tumor necrosis factor-alpha-induced apoptosis in cultured
caspases. J Cell Biol 154:1111 – 6. mouse hepatocytes. Hepatology 36:55 – 64.
256 R. Franco et al.

Nagaoka Y, Iuchi Y, Ikeda Y, Fujii J. 2004. Glutathione reductase is Perez-Cruz I, Carcamo JM, Golde DW. 2003. Vitamin C inhibits
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

expressed at high levels in pancreatic islet cells. Redox Rep FAS-induced apoptosis in monocytes and U937 cells. Blood
9:321 – 4. 102:336 – 43.
Ng KH, Lim BG, Wong KP. 2003. Sulfate conjugating and Perez-Tomas R. 2006. Multidrug resistance: retrospect and
transport functions of MDCK distal tubular cells. Kidney Int prospects in anti-cancer drug treatment. Curr Med Chem
63:976 – 86. 13:1859 – 76.
Ning J, Grant MH. 2000. The role of reduced glutathione and Piga R, Saito Y, Yoshida Y, Niki E. 2007. Cytotoxic effects
glutathione reductase in the cytotoxicity of chromium (VI) in of various stressors on PC12 cells: Involvement of oxida-
osteoblasts. Toxicol In Vitro 14:329 – 35. tive stress and effect of antioxidants. Neurotoxicology 28:
Niwa T, Naito C, Mawjood AH, Imai K. 2000. Increased 67 – 75.
glutathionyl hemoglobin in diabetes mellitus and hyperlipide- Pompella A, Visvikis A, Paolicchi A, De Tata V, Casini AF. 2003.
mia demonstrated by liquid chromatography/electrospray The changing faces of glutathione, a cellular protagonist.
ionization-mass spectrometry. Clin Chem 46:82 – 8. Biochem Pharmacol 66:1499 – 503.
Njalsson R. 2005. Glutathione synthetase deficiency. Cell Mol Life Rahman I. 2005. Regulation of glutathione in inflammation and
Sci 62:1938 – 45. chronic lung diseases. Mutat Res 579:58 – 80.
Njalsson R, Norgren S. 2005. Physiological and pathological Rahman I, Biswas SK, Jimenez LA, Torres M, Forman HJ. 2005.
aspects of GSH metabolism. Acta Paediatr 94:132 – 7. Glutathione, stress responses, and redox signaling in lung
Njalsson R, Ristoff E, Carlsson K, Winkler A, Larsson A, inflammation. Antioxid Redox Signal 7:42 – 59.
Norgren S. 2005. Genotype, enzyme activity, glutathione level, Rana S, Dringen R. 2007. Gap junction hemichannel-mediated
and clinical phenotype in patients with glutathione synthetase release of glutathione from cultured rat astrocytes. Neurosci
deficiency. Hum Genet 116:384 – 9. Lett 415:45 – 8.
Nomura K, Imai H, Koumura T, Nakagawa Y. 2001. Involvement of Raza H, Ahmed I, John A. 2004. Tissue specific expression
mitochondrial phospholipid hydroperoxide glutathione and immunohistochemical localization of glutathione
peroxidase as an antiapoptotic factor. Biol Signals Recept S-transferase in streptozotocin induced diabetic rats: modula-
10:81 – 92. tion by Momordica charantia (karela) extract. Life Sci
O’Donovan DJ, Fernandes CJ. 2000. Mitochondrial glutathione 74:1503 – 11.
and oxidative stress: implications for pulmonary oxygen Raza H, John A. 2004. Glutathione metabolism and oxida-
toxicity in premature infants. Mol Genet Metab 71:352 – 8. tive stress in neonatal rat tissues from streptozotocin-
Obrosova IG, Fathallah L, Greene DA. 2000. Early changes in induced diabetic mothers. Diabetes Metab Res Rev 20:
lipid peroxidation and antioxidative defense in diabetic rat 72 – 8.
retina: effect of DL-alpha-lipoic acid. Eur J Pharmacol Reid M, Jahoor F. 2001. Glutathione in disease. Curr Opin Clin
398:139 – 46. Nutr Metab Care 4:65 – 71.
Obrosova IG, Fathallah L, Lang HJ. 1999. Interaction between Reinehr R, Becker S, Eberle A, Grether-Beck S, Haussinger D.
osmotic and oxidative stress in diabetic precataractous lens: 2005. Involvement of NADPH oxidase isoforms and Src family
studies with a sorbitol dehydrogenase inhibitor. Biochem kinases in CD95-dependent hepatocyte apoptosis. J Biol Chem
Pharmacol 58:1945 – 54. 280:27179 – 94.
Oh SH, Lim SC. 2006. A rapid and transient ROS generation by Ristoff E, Larsson A. 1998. Patients with genetic defects in
cadmium triggers apoptosis via caspase-dependent pathway in the gamma-glutamyl cycle. Chem Biol Interact 111-112:
HepG2 cells and this is inhibited through N-acetylcysteine- 113 – 21.
mediated catalase upregulation. Toxicol Appl Pharmacol Robertson RP. 2004. Chronic oxidative stress as a central
212:212 – 23. mechanism for glucose toxicity in pancreatic islet beta cells in
Opara EC. 2004. Role of oxidative stress in the etiology of type 2 diabetes. J Biol Chem 279:42351 – 4.
diabetes and the effect of antioxidant supplementation on Robertson RP, Harmon J, Tran PO, Poitout V. 2004. Beta-cell
glycemic control. J Investig Med 52:19 – 23. glucose toxicity, lipotoxicity, and chronic oxidative stress in
Pan S, Berk BC. 2007. Glutathiolation regulates tumor necrosis type 2 diabetes. Diabetes 53 Suppl 1:S119 – 24.
factor-alpha-induced caspase-3 cleavage and apoptosis: key Robertson RP, Harmon J, Tran PO, Tanaka Y, Takahashi H.
role for glutaredoxin in the death pathway. Circ Res 100: 2003. Glucose toxicity in beta-cells: type 2 diabetes, good
213 – 9. radicals gone bad, and the glutathione connection. Diabetes
Paolicchi A, Dominici S, Pieri L, Maellaro E, Pompella A. 2002. 52:581 – 7.
Glutathione catabolism as a signaling mechanism. Biochem Rolo AP, Palmeira CM. 2006. Diabetes and mitochondrial
Pharmacol 64:1027 – 35. function: role of hyperglycemia and oxidative stress. Toxicol
Paolisso G, Di Maro G, Pizza G, D’Amore A, Sgambato S, Appl Pharmacol 212:167 – 78.
Tesauro P, Varricchio M, D’Onofrio F. 1992. Plasma Rosado JO, Salvador M, Bonatto D. 2007. Importance of the
GSH/GSSG affects glucose homeostasis in healthy subjects trans-sulfuration pathway in cancer prevention and promotion.
and non-insulin-dependent diabetics. Am J Physiol 263: Mol Cell Biochem 301:1 – 12.
E435 – 40. Rosati E, Sabatini R, Ayroldi E, Tabilio A, Bartoli A, Bruscoli S,
Parcell S. 2002. Sulfur in human nutrition and applications in Simoncelli C, Rossi R, Marconi P. 2004. Apoptosis of human
medicine. Altern Med Rev 7:22 – 44. primary B lymphocytes is inhibited by N-acetyl-L-cysteine.
Park SA, Choi KS, Bang JH, Huh K, Kim SU. 2000. Cisplatin- J Leukoc Biol 76:152 – 61.
induced apoptotic cell death in mouse hybrid neurons is Sakamaki H, Akazawa S, Ishibashi M, Izumino K, Takino H,
blocked by antioxidants through suppression of cisplatin- Yamasaki H, Yamaguchi Y, Goto S, Urata Y, Kondo T, et al.
mediated accumulation of p53 but not of Fas/Fas ligand. 1999. Significance of glutathione-dependent antioxidant sys-
J Neurochem 75:946 – 53. tem in diabetes-induced embryonic malformations. Diabetes
Patriarca S, Furfaro AL, Domenicotti C, Odetti P, Cottalasso D, 48:1138 – 44.
Marinari UM, Pronzato MA, Traverso N. 2005. Supplementa- Sampathkumar R, Balasubramanyam M, Sudarslal S, Rema M,
tion with N-acetylcysteine and taurine failed to restore Mohan V, Balaram P. 2005. Increased glutathionylated
glutathione content in liver of streptozotocin-induced diabetics hemoglobin (HbSSG) in type 2 diabetes subjects with
rats but protected from oxidative stress. Biochim Biophys Acta microangiopathy. Clin Biochem 38:892 – 9.
1741:48 – 54. Santra A, Chowdhury A, Ghatak S, Biswas A, Dhali GK. 2007.
Pennathur S, Heinecke JW. 2004. Mechanisms of oxidative stress Arsenic induces apoptosis in mouse liver is mitochondria
in diabetes: implications for the pathogenesis of vascular dependent and is abrogated by N-acetylcysteine. Toxicol Appl
disease and antioxidant therapy. Front Biosci 9:565 – 74. Pharmacol 220:146 – 55.
GSH in human health and disease 257

Sato T, Machida T, Takahashi S, Iyama S, Sato Y, Kuribayashi K, Valencia E, Hardy G. 2002. Practicalities of glutathione supple-
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

Takada K, Oku T, Kawano Y, Okamoto T, et al. 2004. Fas- mentation in nutritional support. Curr Opin Clin Nutr Metab
mediated apoptosome formation is dependent on reactive Care 5:321 – 6.
oxygen species derived from mitochondrial permeability Valencia E, Hardy G, Marin A. 2001a. Glutathione—nutritional
transition in Jurkat cells. J Immunol 173:285 – 96. and pharmacologic viewpoints: Part V. Nutrition 17:978.
Schafer FQ, Buettner GR. 2001. Redox environment of the Valencia E, Hardy G, Marin A. 2002. Glutathione—nutritional
cell as viewed through the redox state of the glutathione and pharmacologic viewpoints: Part VI. Nutrition 18:
disulfide/glutathione couple. Free Radic Biol Med 30:1191 – 291 – 2.
212. Valencia E, Marin A, Hardy G. 2001b. Glutathione—nutritional
Schulman JD, Goodman SI, Mace JW, Patrick AD, Tietze F, and pharmacological viewpoints: Part II. Nutrition 17:
Butler EJ. 1975. Glutathionuria: inborn error of metabolism 485 – 6.
due to tissue deficiency of gamma-glutamyl transpeptidase. Valencia E, Marin A, Hardy G. 2001c. Glutathione—nutritional
Biochem Biophys Res Commun 65:68 – 74. and pharmacological viewpoints: Part III. Nutrition 17:696 – 7.
Shackelford RE, Heinloth AN, Heard SC, Paules RS. 2005. Valencia E, Marin A, Hardy G. 2001d. Glutathione—nutritional
Cellular and molecular targets of protein S-glutathiolation. and pharmacologic viewpoints: Part I. Nutrition 17:428 – 9.
Antioxid Redox Signal 7:940 – 50. Valencia E, Marin A, Hardy G. 2001e. Glutathione—nutritional
Sharma A, Kharb S, Chugh SN, Kakkar R, Singh GP. 2000. and pharmacologic viewpoints: Part IV. Nutrition 17:783 – 4.
Evaluation of oxidative stress before and after control of Valko M, Leibfritz D, Moncol J, Cronin MT, Mazur M, Telser J.
glycemia and after vitamin E supplementation in diabetic 2007. Free radicals and antioxidants in normal physiological
patients. Metabolism 49:160 – 2. functions and human disease. Int J Biochem Cell Biol 39:
Sharma R, Yang Y, Sharma A, Awasthi S, Awasthi YC. 2004. 44 – 84.
Antioxidant role of glutathione S-transferases: protection Valko M, Morris H, Cronin MT. 2005. Metals, toxicity and
against oxidant toxicity and regulation of stress-mediated oxidative stress. Curr Med Chem 12:1161 – 208.
apoptosis. Antioxid Redox Signal 6:289 – 300. Valko M, Rhodes CJ, Moncol J, Izakovic M, Mazur M. 2006. Free
Shimizu S, Eguchi Y, Kosaka H, Kamiike W, Matsuda H, radicals, metals and antioxidants in oxidative stress-induced
Tsujimoto Y. 1995. Prevention of hypoxia-induced cell death cancer. Chem Biol Interact 160:1 – 40.
by Bcl-2 and Bcl-xL. Nature 374:811 – 13. Valverde M, Rojas E, Kala SV, Kala G, Lieberman MW. 2006.
Sies H. 1999. Glutathione and its role in cellular functions. Free Survival and cell death in cells constitutively unable to
Radic Biol Med 27:916 – 21. synthesize glutathione. Mutat Res 594:172 – 80.
Sindhu RK, Koo JR, Roberts CK, Vaziri ND. 2004. Dysregulation van Bladeren PJ. 2000. Glutathione conjugation as a bioactivation
of hepatic superoxide dismutase, catalase and glutathione reaction. Chem Biol Interact 129:61 – 76.
peroxidase in diabetes: response to insulin and antioxidant van den Dobbelsteen DJ, Nobel CS, Schlegel J, Cotgreave IA,
therapies. Clin Exp Hypertens 26:43 – 53. Orrenius S, Slater AF. 1996. Rapid and specific efflux of
Smith CV, Jones DP, Guenthner TM, Lash LH, Lauterburg BH. reduced glutathione during apoptosis induced by anti-Fas/
1996. Compartmentation of glutathione: implications for the APO-1 antibody. J Biol Chem 271:15420 – 7.
study of toxicity and disease. Toxicol Appl Pharmacol 140: Vander Jagt DL, Hunsaker LA, Vander Jagt TJ, Gomez MS,
1 – 12. Gonzales DM, Deck LM, Royer RE. 1997. Inactivation of
Sze G, Kaplowitz N, Ookhtens M, Lu SC. 1993. Bidirectional glutathione reductase by 4-hydroxynonenal and other endo-
membrane transport of intact glutathione in Hep G2 cells. Am genous aldehydes. Biochem Pharmacol 53:1133 – 40.
J Physiol 265:G1128 – 34. Varghese J, Khandre NS, Sarin A. 2003. Caspase-3 activation is an
Tanaka Y, Tran PO, Harmon J, Robertson RP. 2002. A role for early event and initiates apoptotic damage in a human leukemia
glutathione peroxidase in protecting pancreatic beta cells cell line. Apoptosis 8:363 – 70.
against oxidative stress in a model of glucose toxicity. Proc Wang G, Zhang L, Li Q. 2006. Genetic polymorphisms of
Natl Acad Sci U S A 99:12363 – 8. GSTT1, GSTM1, and NQO1 genes and diabetes mellitus risk
Tanriverdi T, Hanimoglu H, Kacira T, Sanus GZ, Kemerdere R, in Chinese population. Biochem Biophys Res Commun
Atukeren P, Gumustas K, Canbaz B, Kaynar MY. 2007. 341:310 – 3.
Glutathione peroxidase, glutathione reductase and protein Wang W, Ballatori N. 1998. Endogenous glutathione conjugates:
oxidation in patients with glioblastoma multiforme and transi- occurrence and biological functions. Pharmacol Rev 50:
tional meningioma. J Cancer Res Clin Oncol 133:627 – 33. 335 – 56.
Townsend DM, Findlay VL, Tew KD. 2005. Glutathione Watson RW, Rotstein OD, Jimenez M, Parodo J, Marshall JC.
S-transferases as regulators of kinase pathways and anticancer 1997. Augmented intracellular glutathione inhibits Fas-trig-
drug targets. Methods Enzymol 401:287 – 307. gered apoptosis of activated human neutrophils. Blood
Townsend DM, Tew KD, Tapiero H. 2003. The importance of 89:4175 – 81.
glutathione in human disease. Biomed Pharmacother 57: Wernerman J, Hammarqvist F. 1999. Modulation of endogenous
145 – 55. glutathione availability. Curr Opin Clin Nutr Metab Care
Traverso N, Menini S, Odetti P, Pronzato MA, Cottalasso D, 2:487 – 92.
Marinari UM. 2002. Diabetes impairs the enzymatic disposal Winiarska K, Drozak J, Wegrzynowicz M, Fraczyk T, Bryla J.
of 4-hydroxynonenal in rat liver. Free Radic Biol Med 32: 2004. Diabetes-induced changes in glucose synthesis, intracel-
350 – 9. lular glutathione status and hydroxyl free radical generation in
Ueda S, Nakamura H, Masutani H, Sasada T, Yonehara S, rabbit kidney-cortex tubules. Mol Cell Biochem 261:91 – 8.
Takabayashi A, Yamaoka Y, Yodoi J. 1998. Redox regulation Wlodek L. 2002. Beneficial and harmful effects of thiols. Pol J
of caspase-3(-like) protease activity: regulatory roles of thior- Pharmacol 54:215 – 23.
edoxin and cytochrome c. J Immunol 161:6689 – 95. Wu G, Fang YZ, Yang S, Lupton JR, Turner ND. 2004.
Ulusu NN, Sahilli M, Avci A, Canbolat O, Ozansoy G, Ari N, Glutathione metabolism and its implications for health. J Nutr
Bali M, Stefek M, Stolc S, Gajdosik A, et al. 2003. Pentose 134:489 – 92.
phosphate pathway, glutathione-dependent enzymes and anti- Xu L, Koumenis IL, Tilly JL, Giffard RG. 1999. Overexpression
oxidant defense during oxidative stress in diabetic rodent brain of bcl-xL protects astrocytes from glucose deprivation and is
and peripheral organs: effects of stobadine and vitamin E. associated with higher glutathione, ferritin, and iron levels.
Neurochem Res 28:815 – 23. Anesthesiology 91:1036 – 46.
Um HD, Orenstein JM, Wahl SM. 1996. Fas mediates apoptosis Xu Z, Patel KP, Lou MF, Rozanski GJ. 2002. Up-regulation of
in human monocytes by a reactive oxygen intermediate K(þ) channels in diabetic rat ventricular myocytes by insulin
dependent pathway. J Immunol 156:3469 – 77. and glutathione. Cardiovasc Res 53:80 – 8.
258 R. Franco et al.

Yadav S, Zajac E, Singhal SS, Awasthi S. 2007. Linking stress- Yoshida K, Hirokawa J, Tagami S, Kawakami Y, Urata Y,
Downloaded By: [University of Nevada Reno] At: 23:49 21 April 2008

signaling, glutathione metabolism, signaling pathways and Kondo T. 1995. Weakened cellular scavenging activity against
xenobiotic transporters. Cancer Metastasis Rev 26: oxidative stress in diabetes mellitus: regulation of glutathione
59 – 69. synthesis and efflux. Diabetologia 38:201 – 10.
Yalin S, Hatungil R, Tamer L, Ates NA, Dogruer N, Yildirim H, Yue KK, Chung WS, Leung AW, Cheng CH. 2003. Redox
Karakas S, Atik U. 2006. Glutathione S-transferase gene changes precede the occurrence of oxidative stress in eyes
polymorphisms in Turkish patients with diabetes mellitus. Cell and aorta, but not in kidneys of diabetic rats. Life Sci 73:
Biochem Funct 25:509 – 13. 2557 – 70.
Yang MS, Chan HW, Yu LC. 2006a. Glutathione peroxidase and Zech B, Wilm M, van Eldik R, Brune B. 1999. Mass spectrometric
glutathione reductase activities are partially responsible for analysis of nitric oxide-modified caspase-3. J Biol Chem
determining the susceptibility of cells to oxidative stress. 274:20931 – 6.
Toxicology 226:126 – 30. Zhai Q, Ji H, Zheng Z, Yu X, Sun L, Liu X. 2000. Copper induces
Yang P, Ebbert JO, Sun Z, Weinshilboum RM. 2006b. Role of the apoptosis in BA/F3beta cells: Bax, reactive oxygen species, and
glutathione metabolic pathway in lung cancer treatment and NFkappaB are involved. J Cell Physiol 184:161 – 70.
prognosis: a review. J Clin Oncol 24:1761 – 9. Zhou L, Jing Y, Styblo M, Chen Z, Waxman S. 2005.
Yang S, Zhu H, Li Y, Lin H, Gabrielson K, Trush MA, Diehl AM. Glutathione-S-transferase pi inhibits As2O3-induced apoptosis
2000. Mitochondrial adaptations to obesity-related oxidant in lymphoma cells: involvement of hydrogen peroxide catabo-
stress. Arch Biochem Biophys 378:259 – 68. lism. Blood 105:1198 – 203.
Yasukawa T, Tokunaga E, Ota H, Sugita H, Martyn JA,
Kaneki M. 2005. S-nitrosylation-dependent inactivation of
Akt/protein kinase B in insulin resistance. J Biol Chem
280:7511 – 18.

View publication stats

You might also like