You are on page 1of 9

Toxicology and Applied Pharmacology 204 (2005) 320 – 328

www.elsevier.com/locate/ytaap

Review

Intestinal glutathione: determinant of mucosal peroxide transport,


metabolism, and oxidative susceptibility
Tak Yee Aw*
Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center,
1501 Kings Highway, Shreveport, LA 71130-3932, USA

Received 8 October 2004; accepted 15 November 2004


Available online 23 January 2005

Abstract

The intestine is a primary site of nutrient absorption and a critical defense barrier against dietary-derived mutagens, carcinogens, and
oxidants. Accumulation of oxidants like peroxidized lipids in the gut lumen can contribute to impairment of mucosal metabolic pathways,
enterocyte dysfunction independent of cell injury, and development of gut pathologies, such as inflammation and cancer. Despite this
recognition, we know little of the pathways of intestinal transport, metabolism, and luminal disposition of dietary peroxides in vivo or of the
underlying mechanisms of lipid peroxide-induced genesis of intestinal disease processes. This chapter summarizes our current understanding
of the determinants of intestinal absorption and metabolism of peroxidized lipids. I will review experimental evidence from our laboratory
and others (Table 1) supporting the pivotal role that glutathione (GSH) and reduced nicotinamide adenine dinucleotide phosphate (NADPH)
play in mucosal transport and metabolism of lipid hydroperoxides and how reductant availability can be compromised under chronic stress
such as hypoxia, and the influence of GSH on oxidative susceptibility, and redox contribution to genesis of gut disorders. The discussion is
pertinent to understanding dietary lipid peroxides and GSH redox balance in intestinal physiology and pathophysiology and the significance
of luminal GSH in preserving the integrity of the intestinal epithelium.
D 2004 Elsevier Inc. All rights reserved.

Keywords: Intestinal peroxide transport and disposition; Glutathione and intestinal peroxide elimination; Lymphatic peroxide transport; Intestinal apoptosis;
Mitochondrial apoptotic signaling; Oxidative stress and apoptosis; Intestinal redox and proliferation; GSH/GSSG redox and apoptosis; Glutathione and
intestinal redox balance

Contents

Glutathione and intestinal disposition of peroxidized lipids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 321


Lipid peroxides and intestinal pathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 321
GSH and intestinal lipid peroxide absorption, metabolism, and lymphatic transport . . . . . . . . . . . . . . . . . . . . . . 321
Intestinal handling of luminal lipid peroxides. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 321
GSH in intestinal peroxide transport and elimination . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 322
NADPH and GSH redox cycle in intestinal hydroperoxide metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . 324
Intestinal GSH/GSSG redox balance, oxidative susceptibility, and gut pathology . . . . . . . . . . . . . . . . . . . . . . . . . 325
Cellular GSH redox governs intestinal cell transition and oxidative vulnerability . . . . . . . . . . . . . . . . . . . . . . . 325
GSH redox imbalance and development of intestinal disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 326
Summary and perspective . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 326
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 327
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 327

* Fax: +1 318 475 4217.


E-mail address: taw@lsuhsc.edu.

0041-008X/$ - see front matter D 2004 Elsevier Inc. All rights reserved.
doi:10.1016/j.taap.2004.11.016
T.Y. Aw / Toxicology and Applied Pharmacology 204 (2005) 320–328 321

Glutathione and intestinal disposition of peroxidized peroxides or their metabolic products into lymph in association
lipids with lipoproteins. Early studies in the 1960s and 1970s have
investigated the absorption and lymphatic transport of fatty
Lipid peroxides and intestinal pathology acid hydroperoxides (Bergen and Draper, 1970; Bunyan et al.,
1968; Glavind and Sylven, 1970; Nagatsugawa and Kaneda,
Lipid peroxides are a major source of dietary oxidants 1983), but results from these studies are equivocal regarding
of mutagenic or carcinogenic potential that are of nutri- whether luminal peroxidized lipids are, in fact, absorbed and
tional and toxicological importance (Ames, 1983; Kinlen, processed by the small intestine. The difficulty is due, in part,
1983). By virtue of their ability to generate oxyradicals, to different experimental models that have been employed to
lipid peroxides are capable of initiating degenerative study intestinal hydroperoxide disposition under in vitro and in
processes and promote digestive system disorders, such vivo conditions. An experimental model of some utility for
as inflammation and cancer (Grisham and Granger, 1998; addressing the issue of intestinal handling of lipid peroxides is
Parks et al., 1983). Dietary intake of highly unsaturated that of the everted intestinal sac. First described in 1954 by
fats is an important contributor to lipid peroxides in the Wilson and Wiseman, the intestinal everted sac provided a
intestinal lumen, and an estimated daily intake of 1.4 convenient in vitro method for quantifying transport of
mmol lipid peroxides per day could occur in humans nutrients when precautions were taken to maintain mucosal
consuming an average of 84 g of fat daily (Wolff and blood flow and oxygen delivery through minimizing villus
Nourooz-Zadeh, 1996). In animal studies, luminal peroxide vasoconstriction (Pritchard and Porteous, 1977; Wilson and
contents of 0.4 Amol have been detected after experimental Wiseman, 1954). The subsequent establishment of the lymph
duodenal infusion of peroxidized lipids (Aw et al., 1992); fistula rat model (see below) offers better quantitative
levels that have been shown to alter intestinal cell measures of the absorption and metabolism of peroxidized
responses in vitro (0.2–10 AM; Gotoh et al., 2002; Wang lipids without the issue of tissue oxygenation associated with
et al., 2000). Early studies have demonstrated that the the everted sac model.
toxicity of dietary polyunsaturated oils correlated directly Evidence for transport of dietary lipid peroxides into the
with their peroxide contents and that excessive consump- circulation is equally controversial. A few studies docu-
tion of lipid peroxides was cytotoxic in vivo (Andrews et mented the presence of lipid peroxides in the lymph
al., 1960; Kaneda et al., 1955; Kimura et al., 1984). (Nagatsugawa and Kaneda, 1983), isolated chylomicrons
Although luminal toxic concentrations have not been (Staprans et al., 1994), blood plasma (Ursini et al., 1998),
demonstrated in humans, the intake of polysaturated fats and tissues (Bunyan et al., 1968; Kaneda et al., 1955) after
can elevate peroxide contents to levels that impact intestinal administration of lipid peroxide containing diet. Others were
integrity by induction of tissue oxidative stress and redox unable to document the presence of lipid peroxides in lymph
imbalance. In rats, administration of hydroperoxy and (Andrews et al., 1960; Bergen and Draper, 1970; Glavind
hydroxy fatty acids stimulated DNA synthesis and induced and Sylven, 1970; Glavind et al., 1971); rather, evidence
ornithine decarboxylase activity consistent with enhanced supports the absorption of degradation products of lipid
cellular proliferation (Bull et al., 1984). Moreover, mucosal peroxides, such as hydroxyl fatty acids, conjugated dienes,
hypertrophy of the colon developed in rats given peroxi- and aldehydes (Andrews et al., 1960; Bergen and Draper,
dized ethyl linoleate (Hara et al., 1996). These results 1970; Glavind and Sylven, 1970; Glavind et al., 1971;
underscore the deleterious impact that lipid peroxides have Kanazawa and Ashida, 1998; Kanazawa et al., 1985; Orada
on normal intestinal cell turnover processes and highlight et al., 1987). The reasons for the different conflicting
their tumorigenic potential. Indeed, the increased incidence observations are unclear; notably, the factors that affect lipid
of colon cancer with high intake of dietary fats (Carroll and peroxide absorption, metabolism, and lymphatic transport
Khor, 1975; Correa et al., 1982; Reddy, 1983) is consistent were not evaluated in these studies. The presence of anti-
with the paradigm that dietary fat is a risk factor for colon oxidant defenses in mesenteric lymph offers one possible
cancer development. Despite this recognition, the disposi- mechanism that accounts for the reduced presence of
tion of luminal peroxidized lipids by the small intestine lymphatic lipid peroxides observed after intragastric admin-
remains poorly understood. istration of vegetable oil containing triglyceride hydro-
peroxides (Mohr et al., 1999). Additionally, the appearance
GSH and intestinal lipid peroxide absorption, metabolism, of hydroxyl compounds that are reduction products of lipid
and lymphatic transport peroxides in mesenteric lymph points to a role for mucosal
glutathione (GSH) and GSH peroxidases in enterocyte
Intestinal handling of luminal lipid peroxides metabolism of luminal lipid peroxides. Thus, differences
The overall processing of luminal lipid peroxides by the in mucosal GSH status under the various dietary and
small intestine can be considered to occur in three phases. The experimental conditions (see below) could conceivably
respective absorptive, metabolic, and transport phases involve explain the lack of general agreement in the literature on
the uptake of peroxides from the lumen, the intracellular how luminal peroxides are handled by the small intestine
catabolism of the absorbed peroxides, and finally the release of (Table 1).
322 T.Y. Aw / Toxicology and Applied Pharmacology 204 (2005) 320–328

Table 1 GSH intake and biliary GSH output are the major sources of
Summary of literature evidence for glutathione as a determinant of mucosal luminal GSH. The human diet varies considerably in GSH
peroxide transport, metabolism, and oxidative susceptibility
levels (Jones et al., 1992; Wierzbicka et al., 1989), and ~50%
Studies Literature references of total hepatic GSH efflux goes into bile (Aw, 1994).
Intestinal peroxide handling Furthermore, oral GSH administration was shown to prevent
Peroxide absorption/lymphatic Bergen and Draper (1970), Bunyan et al.
severe degeneration of jejunal and colonic epithelial cells
transport (1968), Glavind and Sylven (1970),
Glavind et al. (1971), Kanazawa and induced by chronic GSH deficiency (Martensson et al.,
Ashida (1998), Mohr et al. (1999), 1990), consistent with its physiological role in preservation of
Nagatsugawa and Kaneda (1983), Orada the integrity of the intestinal epithelium.
et al. (1987), Staprans et al. (1994), Evidence to date implicates GSH as a key determinant in
Ursini et al. (1998)
the elimination of peroxides by the intestine (Aw and
GSH in peroxide absorption Aw (1994), Aw and Williams (1992),
and lymphatic transport Aw et al. (1992), Kowalski et al. (1990) Williams, 1992; Aw et al., 1992; Kowalski et al., 1990).
NADPH and GSH redox Using our conscious lymph fistula rat model, we have
cycle in mucosal provided novel insights into the driving force for lipid
peroxide metabolism peroxide absorption and removal by the small intestine (Aw et
Control conditions Aw and Rhoads (1994), Tribble and
al., 1992). Conceptually, this model predicts that at a given
Jones (1990), Williams and Aw (1994),
Wingler et al. (2000) peroxide concentration, the amount of peroxide (designated
Pathophysiological stress Chu et al. (1993), Iwakiri et al. (1995), as ROOH in Fig. 1) recovered from the intestinal lumen and
LeGrand and Aw (1996, 1997, 1998), lymph is governed by the mucosal GSH status. During GSH
Reddy and Tappel (1974), Vilas et al. sufficiency, increased intracellular peroxide metabolism
(1976)
drives mucosal ROOH uptake that results in decreased
Intestinal GSH/GSSG redox and oxidative susceptibility luminal ROOH retention and attenuated ROOH output into
Redox and cell transitions: intestinal lymph (Fig. 1A). This mechanism of GSH-depend-
Cellular models ent bmetabolic trappingQ highly favors peroxide catabolism
Proliferation Gotoh et al. (2002) and offers an efficient mode for rapid peroxide disposal in the
Growth arrest/differentiation Nkabyo et al. (2002), Noda et al. (2001)
small intestine. Conversely, during GSH deficiency,
Apoptosis Cai et al. (2004), Wang et al. (2000)
Susceptibility in nonintestinal Ekshyyan and Aw (2004), Pias and Aw decreased intracellular peroxide catabolism impairs mucosal
cells (2002a, 2002b), Pias et al. (2003) ROOH absorption, increases luminal ROOH retention, and
Redox and intestinal turnover: Iwakiri et al. (2001), Tsunada et al. promotes lymphatic ROOH transport (Fig. 1A). Thus, the
animal models (2003a, 2003b) capacity for and the efficiency of peroxide catabolism within
Redox and intestinal disorders Keshavarzian et al. (1992),
enterocytes largely determine peroxide elimination from the
Simmons et al. (1992),
Tsunada et al. (2003a, 2003b, 2000c) intestinal lumen. Experimentally, the lymph fistula rat model
allows for the simultaneous quantification of peroxide
recovery in intestinal lumen and lymph, and thereby provides
GSH in intestinal peroxide transport and elimination insights into the intestine’s capacity for peroxide metabolism
in vivo (Aw et al., 1992). As illustrated in the schematic
Glutathione (GSH) is a naturally occurring cellular profiles in Fig. 1B based on experimentally generated results,
reductant and antioxidant (Kaplowitz et al., 1985; Kauffman but without showing actual quantitative data, we found that
et al., 1977) that functions to detoxify reactive oxygen increased intestinal peroxide absorption as well as reduced
metabolites of endogenous or exogenous origins. GSH is luminal and lymph recoveries of infused lipid peroxides
present in high concentrations in tissues (Aw et al., 1992; correlate with higher mucosal GSH concentrations (Aw and
Kaplowitz et al., 1985; Shan et al., 1990), and normal tissue Williams, 1992; Aw et al., 1992; LeGrand and Aw, 2001),
GSH homeostasis is maintained by de novo synthesis from suggestive of an overall decrease in peroxide absorption and
cysteine, from regeneration of glutathione disulfide, GSSG, metabolism in association with reduced GSH levels. These
and from GSH uptake of exogenous sources via Na+- results underscore the quantitative importance of mucosal
dependent transport systems. Cellular GSH transport has GSH in the removal of luminal lipid peroxides.
been linked to cytoprotection against peroxide-induced Exogenous GSH supplements either from the diet or bile
damage in a variety of cell types, including enterocytes increases mucosal GSH concentrations and promotes metab-
(Lash et al., 1986), type II alveolar cells (Hagen et al., 1986), olism of lipid peroxides, thereby increasing luminal peroxide
renal proximal tubule cells (Hagen et al., 1988), and uptake into enterocytes (Aw and Williams, 1992; Kowalski et
endothelial cells (Andreoli et al., 1986). Moreover, GSH al., 1990). That biliary GSH is a major contributor to the
has been shown to be transported intact into the small luminal GSH pool (Aw, 1994) is evidenced by studies
intestine in vivo to supplement the intracellular GSH pool showing that diversion of biliary GSH significantly decreased
(Aw and Williams, 1992; Hagen and Jones, 1987; Hagen et intestinal peroxide metabolism that results in elevated
al., 1990; Lash et al., 1986), suggesting that luminal GSH can peroxide recoveries in lumen and lymph (Aw, 1994; LeGrand
contribute to maintaining mucosal thiol balance. Dietary and Aw, 2001). Using a similar schematic profile as that
T.Y. Aw / Toxicology and Applied Pharmacology 204 (2005) 320–328 323

Fig. 1. Panel A represents a working model for absorption and lymphatic transport of luminal lipid peroxides in intestinal cells with GSH-sufficient or GSH-
deficient status. The abbreviations are GSH, GSSG: reduced glutathione and glutathione disulfide; S, D: GSH-sufficient and -deficient status, respectively;
ROOH, ROH: lipid peroxide and hydroxide, respectively; [Lo], [Hi]: low and high peroxide concentrations, respectively. Panel B represents profiles of
intestinal peroxide absorption, luminal peroxide accumulation, and lymphatic peroxide transport as a function of mucosal GSH concentrations. The
enhancement of intracellular lipid peroxide metabolism will accelerate peroxide uptake from the gut lumen resulting in decrease luminal accumulation and
reduce peroxide output into lymph secondary to cellular detoxication. Redrawn from LeGrand and Aw (2001).

Fig. 2. (A) Exogenous GSH enhances intestinal elimination of lipid peroxides. Supplementation of luminal GSH increases mucosal GSH concentration that
enhances luminal peroxide absorption and peroxide metabolism and reduces lymphatic peroxide transport. Dotted arrow 1 shows the direction of change in
intestinal peroxide uptake (upper panel) and lymphatic transport (lower panel) with GSH supplementation. Blockade of intestinal uptake of GSH from the gut
lumen decreases mucosal GSH concentration that impairs luminal peroxide update and peroxide metabolism and enhances lymphatic peroxide output. Dotted
arrow 2 shows the direction of change in peroxide absorption (upper panel) and lymphatic transport (lower panel) when luminal GSH uptake is prevented. The
profiles are based on experimentally generated data from Aw and Williams (1992). Redrawn from LeGrand and Aw, 2001. (B) Diversion of biliary GSH
decreases intestinal elimination of lipid peroxides. Diversion of biliary GSH in bile fistula animals decreases mucosal GSH that compromises intestinal
peroxide uptake and peroxide metabolism, and enhances lymphatic transport. The dashed line (1: bile fistula) shows the profiles for peroxide absorption (upper
panel) and lymphatic transport (lower panel) in bile fistula animals. Exogenous GSH supply in bile fistula animals restores luminal peroxide elimination and
decreases peroxide output into lymph. The solid line 2 represents the profiles for peroxide absorption (upper panel) and lymphatic transport (lower panel) in
GSH-supplemented bile fistula animals, and the dotted arrow 3 shows the direction of change in these parameters following GSH supplementation. The profiles
of peroxide absorption and lymphatic transport are based on experimentally generated data from Aw (1994). Redrawn from LeGrand and Aw (2001).
324 T.Y. Aw / Toxicology and Applied Pharmacology 204 (2005) 320–328

shown in Fig. 1B that is based on experimentally generated NADPH and GSH redox cycle in intestinal hydroperoxide
results (Aw and Williams, 1992), the effect of exogenous metabolism
GSH supplementation on intestinal absorption, intramucosal
metabolism, and lymphatic output of lipid peroxides can be The GSH redox cycle is a key intracellular detoxication
summarized in Fig. 2A. The results show that elevating mechanism for peroxide elimination. Indeed, studies by
luminal GSH leads to an increased mucosal GSH that Wingler et al. (2000) demonstrated that intestinal GSH
promotes luminal peroxide uptake, enhances cellular per- peroxidase plays a key role in attenuating transport of lipid
oxide metabolism, and decreases lymphatic peroxide trans- peroxides and thereby prevent against oxidative damage in a
port (dotted Arrow 1). Conversely, blockade of luminal GSH CaCo-2 cell culture model. GSH peroxidase and GSSG
uptake or utilization leads to decreased mucosal GSH that reductase function as an enzyme pair in the reduction of
attenuates luminal peroxide uptake, compromises cellular peroxides with concomitant oxidation of GSH and the
peroxide metabolism, and promotes lymphatic output (dotted regeneration of GSH with reducing equivalents donated from
arrow 2). The result on the effect of bilary GSH diversion on nicotinamide adenine dinucleotide phosphate (NADPH). The
intestinal handling of lipid peroxides is summarized in the supply of NADPH is functionally coupled to the pentose
schematic model illustrated in Fig. 2B. Again, for simplicity, phosphate pathway (Eggleston and Krebs, 1974) and is
the pattern of changes as based on experimental results (Aw, regulated by glucose availability and glucose 6-phosphate
1994) is shown without presentation of the actual quantitative dehydrogenase activity (Bousignone and Flora, 1972). Given
data. The data show that biliary diversion of GSH via a bile that enterocyte GSH synthetic rate is low (Williams and Aw,
fistula resulted in decreased mucosal GSH (dashed line, 1994) as compared to hepatocytes (Shan et al., 1990), GSSG
2:bile fistula) as compared to controls. The decreased reduction is quantitatively an important mechanism for
mucosal GSH in the combined bile fisula and lymph fistula intestinal GSH maintenance under high rates of lipid peroxide
rats leads to reduced luminal peroxide removal, compromised reduction (Aw and Rhoads, 1994). Glucose stimulation of
cellular peroxide metabolism, and elevated lymphatic output. enterocyte NADPH supply (Aw and Rhoads, 1994) suggests
Notably, supplementation of these animals with exogenous that GSH redox cycle activity is dependent on exogenous
GSH restores mucosal GSH content (solid line, 2:+GSH) that glucose availability for NADPH production. The contention
promoted luminal peroxide uptake, increased cellular per- that constant glucose supply is critical in maintaining
oxide detoxication, and reduced peroxide output into lymph NADPH generation and GSH redox cycle function in vivo
(dotted arrow 3). Collectively, these studies using the lymph is borne out by studies in the lymph fistula rat wherein
fistula rat model without or with bile fistula provide removal of glucose from the lipid peroxide infusate in GSH-
reasonable quantitative measures of the absorption, metabo- sufficient animals results in elevated recoveries of luminal
lism, and lymphatic transport of peroxidized lipids in the and lymph peroxide contents, reminiscent of the GSH-
small intestine under various conditions of mucosal GSH deficient state (Aw and Williams, 1992; Aw et al., 1992).
sufficiency or GSH deficiency. This responsiveness of the gut to glucose suggests that
While current evidence support mucosal GSH and intra- nutrient availability is an important contributing factor in the
cellular metabolism as the major driving force governing intestinal catabolism of luminal peroxides and raises the
intestinal lipid peroxide uptake, the bioavailability of interesting possibility of regulation of mucosal peroxide
peroxidized fatty acids could be influenced by their physical disposition by glucose of luminal or plasma origin. Moreover,
chemical properties given that lipid uptake from a micellar because of the difference in quantitative NADPH supply in
phase is a generally accepted paradigm for intestinal transport the presence of glucose between the intestine and liver (0.5–
of free fatty acids into enterocytes (Johnston and Borgstrom, 1.0 nmol min 1 per 106 cells and 5–8 nmol min 1 per 106
1964). The formation of micelles has been shown to be a cells, respectively; Aw and Rhoads, 1994; Tribble and Jones,
mechanism for overcoming the resistance of the unstirred 1990), a greater sensitivity of the small intestine to peroxide-
water barrier to facilitate rapid transfer of fatty acids to the induced injury is expected.
intestinal cell membrane. At present, the extent to which Interestingly, studies from our laboratory and others show
oxidative modifications of lipids influence their luminal that intestinal GSH peroxidase and GSSG reductase activities
solubility, their degree of penetration of plasma membrane, are minimally modified under conditions of peroxide
and their ability to participate in bile acid mixed micelles are challenge (Reddy and Tappel, 1974; Vilas et al., 1976),
unknown nor is it known whether micellar formation is chronic hypoxia (LeGrand and Aw, 1996), and diabetes
necessary for and critical to mucosal uptake of peroxidized (Iwakiri et al., 1995), suggesting that the redox enzymes per
lipids. Future investigations into the effects of carbon chain se are generally not rate limiting in overall lipid peroxide
lengths, degree of unsaturation, and extent of oxidation of catabolism and removal in the intestine. Notwithstanding, a
polyunsaturated fatty acids on their absorption characteristics role for the specific intestinal isoform of GSH peroxidase
should provide additional insights into how oxidative (GSHPX-GI, Chu et al., 1993) has not been vigorously
modifications of fatty acids influence the mucosal handling evaluated, but given its catalytic rate at one-tenth that of the
of varied luminal lipids of different fatty acid composition classical GSH peroxidase, GSHPX-GI is unlikely to signifi-
and degree of peroxidation. cantly influence peroxide elimination. Rather, literature
T.Y. Aw / Toxicology and Applied Pharmacology 204 (2005) 320–328 325

evidence suggests that it is the efficiency of lipid peroxide and quiescent state typifies fully differentiated tissues like
absorption from the lumen (Aw et al., 1992) and the liver, kidney, brain, and intestine. Shifting cellular control
availability of cellular reductants (GSH and NADPH) (Aw checkpoints toward either GSH/GSSG reduction or oxida-
and Rhoads, 1994; Aw and Williams, 1992; Kowalski et al., tion can elicit phase transition of a cell from a quiescent
1990) that largely dictate the kinetics and extent of intra- state to that of a proliferative, growth arrested, apoptotic, or
cellular peroxide detoxication by the intestine. necrotic state (Aw, 1999; Flores and McCord, 1997).
It is apparent that factors that alter intestinal GSH Depending on cell types, three cellular responses are
concentrations, the supply of NADPH, or the function of possible (Fig. 3). Terminally quiescent or differentiated
the GSH redox cycle are expected to impact metabolic cells such as hepatocytes or neurons generally exhibit a
processing of dietary lipid peroxides. We have demonstrated biological constraint to proliferate (Curve I) and increased
that chronic oxygen deficiency has a profound effect on oxidative stress induces cell apoptosis. Mitotic-competent
intestinal handling of luminal peroxides due in part to a enterocytes can maintain a genetic program to proliferate in
generalized hypoxia-induced decrease in the GSH-to-GSSG response to mitogenic stimuli (Curve II), wherein cells
ratio in the small intestine (LeGrand and Aw, 1996, 1997). In overcome the G0/G1 regulatory checkpoint to enter the S
this state of tissue oxidative stress, GSH-dependent metab- phase. Transformed cells possessing altered regulatory cell
olism of lipid peroxides is compromised, resulting in cycle barriers respond to oxidative challenge with greater
decreased luminal peroxide removal and elevated abluminal proliferation or apoptosis (Curve III). Thus, depending on
peroxide recovery (LeGrand and Aw, 1998). Notably, the the degree and duration of the oxidant challenges or redox
NADPH production rate in hypoxic enterocytes is 50% lower shifts, distinct cell transitions or oxidative susceptibility will
than that in normoxic cells (0.3 versus 0.7 nmol min 1 per be elicited by different cell types.
106 cells), indicating reduced glucose flux through the The fundamental concept that changes in GSH/GSSG
pentose phosphate shunt. This decreased ability of the that modulate intestinal cell proliferation, growth arrest,
hypoxic intestine to maintain GSH redox homeostasis and and apoptosis comes from studies with cultured intestinal
redox cycle activity is likely to favor tissue oxidative cells. We found that treatment of CaCo-2 cells with low
vulnerability and development of gut disorders. concentrations of lipid peroxides (1–5 AM) promotes pro-
liferative activity that corresponds directly to the induction
of mild cellular GSH/GSSG imbalance (Gotoh et al.,
Intestinal GSH/GSSG redox balance, oxidative 2002). As the peroxide induces a progressive oxidized
susceptibility, and gut pathology shift in the GSH/GSSG redox state, cells progress from
proliferation to growth arrest (Gotoh et al., 2002; Noda
Cellular GSH redox governs intestinal cell transition and et al., 2001). When intracellular GSH is compromised, the
oxidative vulnerability redox threshold governing the transition between cell
proliferation and growth arrest is lowered (Noda et al.,
One important impact of an altered cellular GSH/GSSG 2001) such that proliferating cell populations with low
status is the induction of cell transitions (Fig. 3). An arrested baseline cellular GSH are selectively vulnerable to oxidiz-
ing events that result in growth arrest or apoptosis. An
interesting aspect is that the cellular redox potential per se
varies with growth status in that the proliferative phenotype
is associated with a more reduced potential than the
differentiated phenotype (Aw, 2003; Nkabyo et al., 2002).
Elicitation of cell apoptosis has been linked to an
intracellular oxidized redox state; notably apoptotic death
is preceded by an early loss of cellular GSH/GSSG redox
balance that was the primary inciting event of the apoptotic
cascade (Wang et al., 2000). Interestingly, we found that in
a nonintestinal cell line, the PC-12 cells, the induction of
GSH/GSSG redox imbalance independently of ROS for-
mation is sufficient to initiate apoptotic signaling (Pias and
Aw, 2002a, 2002b). Moreover, the relative cellular vulner-
ability to apoptosis during peroxide challenge is directly
linked to the cell’s ability to maintain intracellular redox
balance through enhanced NADPH production (Pias and
Aw, 2002a) and the ability to attenuate mitochondrial ROS
Fig. 3. Cellular responses to oxidation–reduction (redox) status. Curves I,
II, and III represent the responses of terminally differentiated, mitotically production (Pias et al., 2003). While an early loss of
competent, and transformed cell types, respectively. Redrawn from Aw cellular redox balance alone is sufficient to induce cell
(2001). apoptosis in intestinal cells (Wang et al., 2000), it remains
326 T.Y. Aw / Toxicology and Applied Pharmacology 204 (2005) 320–328

to be tested as to whether apoptosis can result from a loss pathologies. Chronic gut pathological states like inflamma-
of cellular redox homeostasis per se without an accom- tory bowel diseases are associated with chronic inflamma-
panying ROS generation. tion of the intestine (Fiocchi, 1998; Grisham and Granger,
A recent emerging paradigm from our studies in PC-12 2000; Powrie, 1995). Damage to the intestinal epithelium
cells is that cell transition states (i.e., proliferative versus resulting from an acute inflammatory response is typically
differentiated) can significantly influence their survival viewed as an event secondary to the primary systematic
outcome. At a given peroxide load, the transition from a inflammatory cascade of neutrophil adhering to vascular
proliferative to a quiescent or differentiated phenotype endothelial cells, disruption of the endothelial barrier, and
confers protection against oxidative challenge that is largely enhanced inflammatory cell infiltration into the intestinal
associated with significant increases in total cellular GSH, interstitium. It has been long appreciated that oxidants
most notably in the mitochondrial GSH pool (Ekshyyan and mediate inflammatory reactions, and pivotal to such
Aw, 2004). Studies are underway to address the issue of inflammatory processes, regardless of stimuli, is the
oxidative susceptibility of intestinal cells as they transition induction of cellular oxidative stress. Previous studies have
from the proliferative to the differentiated phenotype. Cai demonstrated a link between reactive oxygen species and
et al. (2004) recently demonstrated that sodium butyrate- the inflammatory response in vivo in inflammatory bowel
induced terminal differentiation of HT 29 colon carcinoma disease (Keshavarzian et al., 1992; Simmons et al., 1992),
cells is associated with apoptotic death that is tightly linked suggesting a role for GSH/GSSG redox in disease etiology.
to caspase activation, suggesting an enhanced vulnerability Indeed, our recent studies demonstrated that induction
with cell differentiation, but a role for redox status in this of intestinal GSH/GSSG imbalance contributes to the
process has not been established. Taken together, it is development of chronic inflammation of the gut in SCID
evident that the cellular redox state serves as a physiological mice reconstituted with CD4+CD45RBhigh T-lymphocytes.
context for optimization of thiol-disulfide control of signal- It is notable that loss of GSH/GSSG redox balance
ing pathways and gene activation in cell proliferation and temporally preceded tissue hyperplasia, mucosal inflamma-
apoptosis. tion, and symptoms of clinical colitis, indicating that redox
Studies in animal models support a paradigm of redox imbalance is a contributor rather than a consequence of
modulation of oxidative susceptibility of the intestinal disease progression. Restored tissue redox homeostasis and
epithelium in vivo. The intestinal mucosa is a labile organ attenuated colitis manifestation were associated with admin-
system whose epithelium turns over every 4–5 days istration of the thiol antioxidant N-acetylcysteine. Recently,
(Creamer et al., 1961; Miller et al., 1977; Tutton, 1973) we demonstrated a linkage between the oxidation of
through a regulated balance of enterocyte proliferation and mucosal GSH and intestinal inflammation and diseases
apoptosis (Iwakiri et al., 2001). We found that rats placed on activity in human ulcerative colitis (Tsunada et al., 2003c).
chronic lipid peroxide diets exhibited suppressed mucosal Active disease state was resolved with normalization of
proliferative and apoptotic activity in accordance with mucosal redox balance by therapeutic intervention with
oxidized tissue GSH/GSSG (Tsunada et al., 2003a, salazosulphapyridine (Tsunada et al., 2003c), an effective
2003b). This state of cytostasis created by chronic peroxide agent for ulcerative colitis and a free radical scavenger
challenge was ameliorated by supplemental GSH that (Ahnfelt-Ronne et al., 1990).
restored tissue GSH/GSSG redox balance (Tsunada et al.,
2003a). Additionally, small intestinal cytostasis was
reversed by administration of epidermal growth factor Summary and perspective
(Tsunada et al., 2003b). Significantly, in conjunction with
an increase in colonic GSH levels, EGF administration Our current understanding of the physiological deter-
induced a hyperproliferative response in the colon that minants of intestinal disposition of luminal lipid peroxides
normally exhibits a slow cell turnover (Tsunada et al., is limited, despite the recognition that oxidants like
2003b). Taken together, these results demonstrate the peroxidized lipids can contribute to the pathogenesis of
centrality of cellular GSH/GSSG redox in modulating cell degenerative processes in the gut. Studies from our
transitions and their oxidative susceptibility. Thus, physio- laboratory in the conscious lymph fistula rat underscore
logical or pathological factors that induce peroxide gen- a pivotal role for intestinal GSH in quantitative lipid
eration, decrease luminal GSH transport, or promote peroxide transport and metabolism in vivo wherein luminal
intestinal GSH oxidation are expected to impact intestinal peroxide elimination is found to be rate limited by the
turnover characteristics and organ survival. availability of tissue reductants (viz., GSH and NADPH)
rather than the functions of the redox enzymes per se.
GSH redox imbalance and development of intestinal Unresolved oxidative stress coupled to the loss of mucosal
disorders redox homeostasis can induce intestinal cell transition to
proliferative or apoptotic phenotypes. Given that dietary
One deleterious consequence of an altered cellular redox intake of highly unsaturated fats results in luminal
status in the intestine is the potential for genesis of gut peroxide accumulation and altered tissue redox state, these
T.Y. Aw / Toxicology and Applied Pharmacology 204 (2005) 320–328 327

differential cellular responses to GSH/GSSG redox imbal- Cai, J., Chen, Y., Murphy, T.J., Jones, D.P., Sartorelli, A.C., 2004. Role of
ance could contribute to a loss of intestinal integrity and caspase activation in butyrate-induced terminal differentiation of HT29
colon carcinoma cells. Arch. Biochem. Biophys. 15, 119 – 127.
promote pathogenesis of chronic gut disorders like Carroll, K.K., Khor, H.T., 1975. Dietary fat in relation to tumorigenesis.
inflammatory bowel disease. Evidence that the intestinal Prog. Biochem. Pharmacol. 10, 308 – 353.
epithelium is capable of transporting luminal GSH to Chu, F.F., Doroshow, J.H., Esworthy, R.S., 1993. Expression, characte-
augment intracellular peroxide catabolism and the notion rization, and tissue distribution of a new cellular selenium-dependent
glutathione peroxidase, GSHPX-GI. J. Biol. Chem. 268, 2571 – 2576.
that the etiology of chronic gut pathologies may be directly
Correa, P., Strong, J.P., Johnson, W.D., Pizzolato, P., Haenszel, W., 1982.
linked to an early loss of mucosal redox balance suggest Atherosclerosis and polyps of the colon: quantification of precursors of
that dietary strategies for intestinal redox maintenance coronary heart disease and colon cancer. J. Chronic. Dis. 35, 313 – 320.
would be a viable consideration in chronic gut disease Creamer, B., Shorter, R.G., Bamforth, J., 1961. The turn over and shedding
management. of epithelial cells. Gut 2, 110 – 118.
Eggleston, L.V., Krebs, H.A., 1974. Regulation of the pentose phosphate
cycle. Biochem. J. 138, 425 – 435.
Ekshyyan, O., Aw, T.Y., 2004. Decreased susceptibility of differentiated
Acknowledgments PC12 cells to peroxide-induced apoptosis: relationship to cellular redox.
FASEB J. 18 (5), A1193.
Research in the author’s laboratory was supported by Fiocchi, C., 1998. Inflammatory bowel disease: etiology and pathogenesis.
grants from the National Institutes of Health, DK 44510 and Gastroenterology 115, 182 – 205.
Flores, S.C., McCord, J.M., 1997. Redox regulation by the HIV-1 Tat
DK 43785. transcriptional factor. In: Scandalios, J.G. (Ed.), Oxidative Stress and
the Molecular Biology of Antioxidant Defenses. Cold Spring Harbor
Laboratory Press, Cold Spring Harbor, pp. 117 – 138.
References Glavind, J., Sylven, C., 1970. Intestinal absorption and lymphatic transport
of methyl linoleate hydroperoxide and hydroxyoctadecadienoate in the
rat. Acta Chem. Scand. 24, 3723 – 3728.
Ahnfelt-Ronne, I., Nielsen, O.H., Christensen, A., Langholz, E., Glavind, J., Christensen, F., Sylven, C., 1971. Intestinal absorption and in
Binder, V., Riis, P., 1990. Clinical evidence supporting the radical vivo formation of lipoperoxides in vitamin E deficient rats. Acta Chem.
scavenger mechanism of 5-aminosalicylic acid. Gastrotenterology 98, Scand. 25, 3220 – 3226.
1162 – 1169. Gotoh, Y., Noda, T., Iwakiri, R., Fujimoto, K., Rhoads, C.A., Aw, T.Y.,
Ames, B.N., 1983. Dietary carcinogens and anticarcinogens: oxygen 2002. Lipid peroxide-induced redox imbalance differentially mediates
radicals and degenerative diseases. Science 221, 1256 – 1264. CaCo-2 cell proliferation and growth arrest. Cell Proliferation 35,
Andreoli, S.P., Mallett, C.P., Bergstein, J.M., 1986. Role of glutathione in 221 – 235.
protecting endothelial cells against hydrogen peroxide oxidant injury. Grisham, M.B., Granger, D.N., 1998. Neutrophil-mediated mucosal injury:
J. Lab. Clin. Med. 108, 190 – 198. role of reactive oxygen metabolites. Dig. Dis. Sci. 33, 6S – 15S.
Andrews, J.S., Griffith, W.H., Mead, J.F., Stein, R.A., 1960. Toxicity of air- Grisham, M.B., Granger, D.N., 2000. Leukocyte–endothelial cell inter-
oxidized soybean oil. J. Nutr. 70, 199 – 210. actions in inflammatory bowel disease. In: Kirsner, J.B. (Ed.),
Aw, T.Y., 1994. Biliary glutathione promotes the mucosal metabolism of Inflammatory Bowel Disease. WB Saunders, Philadelphia, pp. 55 – 64.
luminal peroxidized lipids by rat small intestine in vivo. J. Clin. Invest. Hagen, T.M., Jones, D.P., 1987. Transepithelial transport of glutathione
94, 1218 – 1225. in vascularly perfused small intestine of rat. Am. J. Physiol. 252,
Aw, T.Y., 1999. Cellular and molecular responses to oxidative stress and G607 – G613.
redox imbalance. Am. J. Clin. Nutr. 70, 557 – 565. Hagen, T.M., Brown, L.A., Jones, D.P., 1986. Protection against paraquat
Aw, T.Y., 2001. Cellular and molecular responses to oxidative stress and induced injury by exogenous GSH in pulmonary alveolar type II cells.
redox imbalance in intestine. In: Yoshikawa, T. (Ed.), Oxidative Stress Biochem. Pharmacol. 35, 4537 – 4542.
and Digestive Diseases. Karger, Basel, pp. 1 – 12. Hagen, T.M., Aw, T.Y., Jones, D.P., 1988. Glutathione uptake and
Aw, T.Y., 2003. Cellular redox: a modulator of intestinal epithelial cell protection against oxidative injury in isolated kidney cells. Kidney
proliferation. News Physiol. Sci. (NIPS) 18, 201 – 204. Int. 34, 74 – 81.
Aw, T.Y., Rhoads, C.A., 1994. Glucose regulation of hydroperoxide Hagen, T.M., Wierzbicka, G.T, Bowman, B.B., Aw, T.Y., Jones, D.P., 1990.
metabolism in rat intestinal cells. J. Clin. Invest. 94, 2426 – 2434. Fate of dietary glutathione: disposition in the gastrointestinal tract. Am.
Aw, T.Y., Williams, M.W., 1992. Intestinal absorption and lymphatic J. Physiol. 259, G530 – G535.
transport of peroxidized lipids in rats: effect of exogenous GSH. Am. J. Hara, H., Miyashita, K., Ito, S., Kasai, T., 1996. Oxidized ethyl linoleate
Physiol. 263, G665 – G672. induces mucosal hypertrophy of the large intestine and affects cecal
Aw, T.Y., Williams, M.W., Gray, L., 1992. Absorption and lymphatic fermentation of dietary fiber in rats. J. Nutr. 126, 800 – 806.
transport of peroxidized lipids by rat small intestine in vivo: role of Iwakiri, R., Rhoads, C.A., Aw, T.Y., 1995. Determinants of hydroperoxide
mucosal GSH. Am. J. Physiol. 262, G99 – G106. detoxification in diabetic rat intestine: effect of insulin and fasting on
Bergen, F.G., Draper, H.H., 1970. Absorption and metabolism of 1-14C- the glutathione redox cycle. Metabolism 44, 1462 – 1468.
methyl linoleate hydroperoxide. Lipids 5, 976 – 982. Iwakiri, R., Gotoh, Y., Noda, T., Sugihara, H., Fujimoto, K., Fuseler, J.W.,
Bousignone, A., Flora, A.D., 1972. Regulatory properties of glucose Aw, T.Y., 2001. Programmed cell death in rat intestine: effect of feeding
6-phosphate dehydrogenase. Curr. Top. Cell Regul. 6, 21 – 62. and fasting. Scan. J. Gastroenterol. 36, 39 – 47.
Bull, A.W., Nigro, N.D., Golembieski, W.A., Crissman, J.D., Marnett, L.J., Johnston, J.M., Borgstrom, B., 1964. The intestinal absorption and
1984. In vivo stimulation of DNA synthesis and induction of ornithine metabolism of micellar solutions of lipids. Biochim. Biophys. Acta 84,
decarboxylase in rat colon by fatty acid hydroperoxides autoxidation 412 – 423.
products of unsaturated fatty acids. Cancer Res. 44, 4924 – 4928. Jones, D.P., Coates, R.J., Flagg, E.W., Eley, J.W., Block, G., Greenberg,
Bunyan, J., Green, J., Murrell, E.A., Diplock, A.T., Cawthorne, M.A., R.S., Gunter, E.W., Jackson, B., 1992. Glutathione in foods listed in the
1968. On postulated peroxidation of unsaturated lipids in the tissues of National Cancer Institute’s Health Habits and History Food Frequency
vitamin-deficient rats. Br. J. Nutr. 22, 97 – 110. Questionnaire. Nutr. Cancer 17, 57 – 75.
328 T.Y. Aw / Toxicology and Applied Pharmacology 204 (2005) 320–328

Kanazawa, K., Ashida, H., 1998. Dietary hydroperoxides of linoleic acid peroxide-induced apoptosis in mitotic competent undifferentiated cells.
decompose to aldehydes in stomach before being absorbed into the Cell Death Differ. 9, 1007 – 1016.
body. Biochm. Biophys. Acta 1393, 349 – 361. Pias, E.K., Ekshyyan, O.Y., Rhoads, C.A., Fuseler, J.W., Harrison, L., Aw,
Kanazawa, K., Kanazawa, E., Natake, M., 1985. Uptake of secondary T.Y., 2003. Differential effects of superoxide dismutase isoform
autoxidation products of linoleic acid by the rat. Lipids 20, 412 – 419. expression on hydroperoxide-induced apoptosis in PC-12 cells. J. Biol.
Kaneda, T., Sakai, H., Ishii, S., 1955. Nutritive value or toxicity of highly Chem. 278, 13294 – 13301.
unsaturated fatty acids. J. Biochem. 42, 561 – 573. Powrie, F., 1995. Cells in inflammatory bowel disease: protective and
Kaplowitz, N., Aw, T.Y., Ookhtens, M., 1985. The regulation of hepatic pathogenic role. Immunity 3, 171 – 174.
glutathione. Annu. Rev. Pharmacol. Toxicol. 25, 715 – 744. Pritchard, P.J., Porteous, J.W., 1977. Steady-state metabolism and
Kauffman, F.C., Evans, R.K., Thurman, R.G., 1977. Alterations in transport of d-glucose by rat small intestine in vitro. Biochem. J.
nicotinamide and adenine nucleotide systems during mixed-function 164, 1 – 14.
oxidation of p-nitroanisole in perfused livers from normal and Reddy, B.S., 1983. Dietary fat and colon cancer. In: Antrup, H., Williams,
phenobarbital-treated rats. Biochem. J. 166, 583 – 592. G.M. (Eds.), Experimental Colon Carcinogenesis. CRC Press, Boca
Keshavarzian, A., Sedghi, S., Kanofsky, J., List, T., Robinson, C., Ibrahim, Raton, pp. 225 – 239.
C., Winship, D., 1992. Excessive production of ROM by inflamed Reddy, K., Tappel, A.L., 1974. Effect of dietary selenium and autooxidized
colon: analysis by chemilumin probe. Gastroenterology 103, 177 – 185. lipids on the glutathione peroxidse system of gastrointestinal tract and
Kimura, T., Iida, K., Takei, Y., 1984. Mechanisms of adverse effect of other tissues in the rat. J. Nutr. 104, 1069 – 1078.
air-oxidized soybean oil-feeding in rats. J. Nutr. Sci. Vitaminol. 30, Shan, X., Aw, T.Y., Jones, D.P., 1990. Glutathione-dependent protection
125 – 133. against oxidative injury. Pharmacol. Ther. 47, 61 – 71.
Kinlen, L.J., 1983. Fat and cancer. Br. Med. J. 286, 1081 – 1082. Simmons, N.J., Allen, R.E., Stevens, T.R.J., Niall, R., Van Someren, M.,
Kowalski, D.P., Feeley, R.M., Jones, D.P., 1990. Use of exogenous Blake, D.R., Rampton, D.S., 1992. Chemiluminescence assay of
glutathione for metabolism of peroxidized methyl linoleate in rat small mucosal ROM in IBD. Gastroenterology 103, 186 – 196.
intestine. J. Nutr. 120, 1115 – 1121. Staprans, I., Rapp, H.J., Pan, X.M., Kim, K.Y., Feingold, K.R., 1994.
Lash, L.H., Hagen, T.M., Jones, D.P., 1986. Exogenous glutathione protects Oxidized lipids in the diet are a source of oxidized lipid in chylomicrons
intestinal epithelial cells from oxidative injury. Proc. Natl. Acad. Sci. of human serum. Arterioscler., Thromb., Vasc. Biol. 14, 1900 – 1905.
U.S.A. 83, 4641 – 4645. Tribble, D.L., Jones, D.P., 1990. Oxygen dependence of oxidative stress:
LeGrand, T.S., Aw, T.Y., 1996. Chronic hypoxia and glutathione-dependent rate of NADPH supply for maintaining the GSH pool during hypoxia.
detoxication in rat small intestine. Am. J. Physiol. 270, G725 – G729. Biochem. Pharmacol. 39, 729 – 736.
LeGrand, T.S., Aw, T.Y., 1997. Chronic hypoxia, glutathione-dependent Tsunada, S., Iwakiri, R., Fujimoto, K., Aw, T.Y., 2003a. Chronic lipid
detoxication, and metabolic instability in rat small intestine. Am. J. hydroperoxide stress suppresses mucosal proliferation in rat intestine:
Physiol. 272, G328 – G334. potentiation of ornithine decarboxylase activity by epidermal growth
LeGrand, T.S., Aw, T.Y., 1998. Chronic hypoxia alters glucose utilization factor. Dig. Dis. Sci. 48, 2333 – 2341.
during GSH-dependent detoxication in rat small intestine. Am. J. Tsunada, S., Iwakiri, R., Noda, T., Fujimoto, K., Fuseler, J.W., Aw, T.Y.,
Physiol. 274, G376 – G384. 2003b. Chronic exposure to subtoxic levels of peroxidized lipids
LeGrand, T.S., Aw, T.Y., 2001. Intestinal absorption of lipid hydro- suppresses mucosal cell turnover in rat small intestine and the reversal
peroxides. In: Mansbach, C., Kuksis, A., Tso, P. (Eds.), Intestinal Lipid by glutathione. Dig. Dis. Sci. 48, 210 – 222.
Metabolism. Plenum Publishing Corporation, New York, pp. 351 – 366. Tsunada, S., Iwakiri, R., Ootani, H., Aw, T.Y., Fujimoto, K., 2003c.
Martensson, J., Jain, A., Meister, A., 1990. Glutathione is required for Redox imbalance in the colonic mucosa of ulcerative colitis. Scand.
intestinal function. Proc. Natl. Acad. Sci. U.S.A. 87, 1715 – 1719. J. Gastroenterol. 38, 1002 – 1003.
Miller, D.L., Hanson, W., Schedl, H.P., Osborne, J.W., 1977. Proliferation Tutton, P.J., 1973. Variations in crypt cell cycle time and mitotic time in the
rate and transit time of mucosal cells in small intestine of the diabetic small intestine of the rat. Virchows Arch. 13, 68 – 78.
rat. Gastroenterology 73, 1326 – 1332. Ursini, F., Zamburlini, A., Cazzolato, G., Maiorino, M., Bon, G.B., Sevanian,
Mohr, D., Umeda, Y., Redgrave, T.G., Stocker, R., 1999. Antioxidant A., 1998. Postprandial plasma lipid hydroperoxides: a possible link bet-
defenses in rat intestine and mesenteric lymph. Redox Rep. 4, ween diet and artherosclerosis. Free Radical Biol. Med. 25, 250 – 252.
79 – 87. Vilas, N.N., Bell, R.R., Draper, H.H., 1976. Influence of dietary peroxides,
Nagatsugawa, K., Kaneda, T., 1983. Absorption and metabolism of selenium, and vitamin E on glutathione peroxidase of the gastro-
methyl linoleate hydroperoxides in rats. J. Jpn. Oil Chem. Soc. 32, intestinal tract. J. Nutr. 106, 589 – 596.
362 – 366. Wang, T.G., Gotoh, Y., Jennings, M.H., Rhoads, C.A., Aw, T.Y., 2000.
Nkabyo, Y.S., Siegler, T.R., Li, H., Watson, W.H., Jones, D.P., 2002. Cellular redox imbalance induced by lipid hydroperoxide promotes
Glutathione and thioredoxin redox during differentiation in a human apoptosis in human colonic CaCo-2 cells. FASEB J. 14, 1567 – 1576.
colon carcinoma (CaCo-2) cell line. Am. J. Physiol.: Gastrointest. Liver Wierzbicka, G.T., Jagen, T.M., Jones, D.P., 1989. Glutathione in food.
283, G1352 – G1359. J. Food Compos. Anal. 2, 327 – 337.
Noda, T., Iwakiri, R., Fujimoto, K., Aw, T.Y., 2001. Induction of mild Williams, A.D., Aw, T.Y., 1994. Glutathione (GSH) supply for lipid
intracellular redox imbalance inhibits proliferation CaCo-2 cells. hydroperoxide metabolism in intestinal epithelial cells. Gastroentero-
FASEB J. 15, 2131 – 2139. logy 106, 1054.
Orada, M., Miyazawa, T., Fujimoto, K., Kaneda, T., 1987. Absorption and Wilson, T.H., Wiseman, G., 1954. The use of sacs of everted small intestine
metabolism of autoxidation products of methyl linoleate in rat liver. for the study of the transference of substances from the mucosal to the
J. Jpn. Soc. Nutr. Food Sci. 40, 117 – 121. serosal surface. J. Physiol. 123, 116 – 125.
Parks, D.A., Bulkley, G.B., Granger, D.N., 1983. Role of oxygen-derived Wingler, K., Muller, C., Schmehl, K., Florian, S., Brigelius-Flohe, R., 2000.
free radicals in digestive tract diseases. Surgery 94, 415 – 422. Gastrointestinal glutathione peroxidase prevents transport of lipid
Pias, E.K., Aw, T.Y., 2002a. Apoptosis in mitotic competent undiffer- hydroperoxides in CaCo-2 cells. Gastroenterology 119, 420 – 430.
entiated cells is induced by redox imbalance independently of reactive Wolff, S.P., Nourooz-Zadeh, J., 1996. Hypothesis: UK consumption of
oxygen species production. FASEB J. 16, 781 – 790. dietary lipid hydroperoxides—A possible contributory factor to athero-
Pias, E.K., Aw, T.Y., 2002b. Early redox imbalance mediates hydro- sclerosis. Atheroslerosis 119, 261 – 263.

You might also like