Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Am J Physiol Cell Physiol 317: C420–C433, 2019.

First published June 19, 2019; doi:10.1152/ajpcell.00141.2019.

REVIEW Metabolism, Oxidative Stress and Cell Signaling

Oxidative stress pathways in pancreatic ␤-cells and insulin-sensitive cells and


tissues: importance to cell metabolism, function, and dysfunction
Philip Newsholme,1 Kevin N. Keane,1 Rodrigo Carlessi,1 and Vinicius Cruzat2
1
School of Pharmacy and Biomedical Sciences, and Curtin Health Innovation Research Institute, Curtin University, Perth,
Western Australia, Australia; and 2Faculty of Health, Torrens University Australia, Melbourne, Victoria, Australia
Submitted 24 April 2019; accepted in final form 12 June 2019

Newsholme P, Keane KN, Carlessi R, Cruzat V. Oxidative stress pathways in


pancreatic ␤-cells and insulin-sensitive cells and tissues: importance to cell metab-
olism, function, and dysfunction. Am J Physiol Cell Physiol 317: C420 –C433,
2019. First published June 19, 2019; doi:10.1152/ajpcell.00141.2019.—It is now
accepted that nutrient abundance in the blood, especially glucose, leads to the
generation of reactive oxygen species (ROS), ultimately leading to increased
oxidative stress in a variety of tissues. In the absence of an appropriate compen-
satory response from antioxidant mechanisms, the cell, or indeed the tissue,
becomes overwhelmed by oxidative stress, leading to the activation of intracellular
stress-associated pathways. Activation of the same or similar pathways also appears
to play a role in mediating insulin resistance, impaired insulin secretion, and late
diabetic complications. The ability of antioxidants to protect against the oxidative
stress induced by hyperglycemia and elevated free fatty acid (FFA) levels in vitro
suggests a causative role of oxidative stress in mediating the latter clinical
conditions. In this review, we describe common biochemical processes associated
with oxidative stress driven by hyperglycemia and/or elevated FFA and the
resulting clinical outcomes: ␤-cell dysfunction and peripheral tissue insulin resis-
tance.
manganese superoxide dismutase; oxygen species; superoxide dismutase-2; uncou-
pling protein

INTRODUCTION tive stress due to enhanced glucose metabolism and associated


mitochondrial ROS production, especially in the pancreatic
Overview of the Development of Type 1 and Type 2 Diabetes ␤-cell but also in many insulin-sensitive tissues (120), nonen-
Type 1 diabetes (T1DM) is characterized by inflammation of zymatic glycation of proteins, and glucose autoxidation (120).
the pancreatic islets, ␤-cell dysfunction, and death, dependent Oxidative stress may overwhelm antioxidant responses, acti-
on proinflammatory cytokine and other mediators released vating stress-sensitive cellular signaling pathways, as de-
from infiltrating immune cells, including powerful free radical scribed below.
species such as superoxide and nitric oxide (NO) (120). The Oxidative Stress and Diabetes: An Overview
immune mediated attack on the ␤-cells is a very specific type
of autoimmunity. In contrast, type 2 diabetes (T2DM) is Although oxygen is responsible for the expansion of life on
characterized by insulin resistance, excessive hepatic glucose Earth, there are two sides to this molecule: it is essential to
production, and decreased insulin secretion. Insulin resistance sustain energy generation in the mitochondria of cells, but it
most often precedes the onset of type 2 diabetes, is present in can be damaging if not fully reduced to water. Oxygen in the
many of the general population, and is multifactorial (56, 93). air is a relatively nonreactive chemical, but when exposed to
high-energy electron-transferring chemical reactions where re-
Reactive oxygen species (ROS) generation is critical to the
duction is incomplete, it can be converted to various highly
dysfunction of the pancreatic ␤-cell and the development of
reactive chemical forms collectively designated ROS. In mam-
insulin resistance (137) and is discussed in detail in this review. malian cells, a delicate balance exists between the generation
Early insulin resistance is initially overcome by hyperinsulin- of reactive oxygen and reactive nitrogen species (ROS and
emia, so normalizing glucose levels in the blood. However, RNS, respectively), and the removal of such molecules through
persistently elevated glucose may result in intracellular oxida- the action of antioxidants, which may be either chemical [e.g.,
glutathione (GSH)] or enzymatic in structure [e.g., catalase
Address for reprint requests and other correspondence: P. Newsholme,
(CAT)]. ROS and RNS are generated as products of normal
School of Pharmacy and Biomedical Sciences, Curtin University, Perth, WA, cell metabolism and respiration and are absolutely required for
Australia, 6102 (e-mail: philip.newsholme@curtin.edu.au). appropriate cellular function. For example, hydrogen peroxide
C420 0363-6143/19 Copyright © 2019 the American Physiological Society http://www.ajpcell.org
Downloaded from journals.physiology.org/journal/ajpcell (189.239.040.188) on May 14, 2022.
OXIDATIVE STRESS, CELL METABOLISM AND FUNCTION C421
(H2O2) is a central redox signaling molecule that can regulate the probability of further electron leak, O2·⫺generation, and the
cell proliferation, migration, and differentiation via an influ- potential pathological consequences that may affect ␤-cell
ence on signal pathway transduction and posttranslational function in terms of reduced insulin secretion or promote ␤-cell
modifications such as sumoylation (166). Depending on the dysfunction and death, reducing overall pancreatic insulin
location of generation of the ROS and RNS, once the endog- output (53), as described further in the following section.
enous ROS/RNS levels exceed the subcellular buffering anti- The role of other nutrients, such as circulating free fatty
oxidant capacity of the cell, the localized excess reactive acids (FFA), which are elevated in obesity and T2DM, on
species alter the function of various biological molecules such mitochondrial ROS and O2·⫺ are less well defined. It is
as proteins, lipids, and DNA through chemical modification reasonable to consider that increased circulating FFA levels
including nitrosylation, peroxidation, and carbonylation, thus will lead to increased uptake by ␤-cells and subsequently used
promoting dysfunction. This is discussed further below in the as a metabolic fuel by the mitochondria via ␤-oxidation. The
sections on Superoxide-Generating Systems in Pancreatic Is- resulting flux to acetyl-CoA will prime the TCA cycle to
lets and Other ROS Species and Their Sources. Ultimately, this produce more NADH and FADH2, which may again lead to
disturbance in redox homeostasis, resulting in oxidative stress, raised O2·⫺ production. It has been reported that palmitate
promotes impaired function of tissues and organs, which leads exposure can increase mitochondrial O2·⫺ levels in the INS-1E
to chronic disorders at the whole body level. While the level of ␤-cell line (101), and oleate promoted mitochondrial H2O2
ROS/RNS can be increased by exogenous sources such as formation in MIN6 ␤-cells, which was likely to be mediated by
excessive dietary nutrients or tobacco smoking, low endoge- the reaction of O2·⫺ produced by complex I and SOD (90). It
nous levels can rapidly accumulate via a reduction in or was also reported that exposure to oleate and palmitate induced
impairment of the antioxidant arsenal of the cell. Therefore, the ETC uncoupling, increased mitochondrial membrane depolar-
relative levels of ROS, RNS, and antagonistic antioxidants are ization, decreased ATP production, and consequently reduced
critical for maintaining cellular redox homeostasis, preserving insulin secretion in several ␤-cell lines and isolated islets (19,
appropriate cell, tissue, and organ function, and therefore 90). However, other studies have detected oleate-induced O2·⫺
preventing various chronic disorders such as diabetes. An in the cytosol of murine islets (91, 92), which was derived from
imbalance in redox homeostasis is particularly damaging to the NADPH oxidase activity (NOX). Palmitate can also activate
pancreatic ␤-cell, as discussed in detail below. NOX in ␤-cells and islets, leading to increased O2·⫺ produc-
tion, and in acute palmitate exposure conditions NOX can
ROS AND ␤-CELL FUNCTION potentiate glucose-stimulated insulin secretion (GSIS) (62,
Superoxide-Generating Systems in Pancreatic Islets 114, 148). Consequently, NOX is a critical source of intracel-
lular O2·⫺, as it can be stimulated by nutrients such as glucose
Mitochondrial respiration consumes molecular oxygen, or saturated fatty acids and key stimulus-secretion coupling
which is the ultimate acceptor of electrons generated by glu- factors [e.g., Ca2⫹ influx and protein kinase C activation
cose and fatty acid catabolism. During the course of normal (121)].
oxidative phosphorylation, up to 4% of all oxygen consumed is In phagocytic immune cells such as macrophages, mono-
converted into the free radical superoxide (O2·⫺) (16). Super- cytes, and neutrophils, NOX functions to transfer electrons
oxide may be converted into other ROS and may react with from NADPH to molecular oxygen to generate O2·⫺ to kill
RNS to generate the highly destructive peroxynitrite molecule invading pathogens (122). The generalized NOX complex
(120). Superoxide molecules are quickly converted to H2O2 by consists of five major components, including membrane-em-
specific isoforms of the enzyme superoxide dismutase (SOD). bedded gp91phox and gp22phox, and cytosolic p40phox, p47phox,
The H2O2 is then either detoxified to H2O and O2 by glutathi- and p67phox (121). In some cases, activity is also regulated by
one peroxidase (GPx, in the mitochondria) or is detoxified by the small G protein Rac (121). Plasma membrane recruitment
catalase in peroxisomes. However, in the presence of higher of the cytosolic components leads to NOX activation, NADPH
concentrations of the transition metals such as Cu2⫹ or Fe2⫹, utilization, and O2·⫺ production. It has been reported that pancre-
H2O2 can be converted to the highly reactive hydroxyl radical atic ␤-cells express several functional isoforms of NADPH oxi-
and OH⫺ (Fenton reaction). dase, and impairment of p47phox translocation to the plasma
The electron transport chain (ETC) is a major source of membrane reduced glucose oxidation, Ca2⫹ flux, and GSIS in
cellular ROS, and O2·⫺ is one of the most abundant, though ␤-cells (115). Due to the activity of NOX in immune cell
short-lived, ROS species in cells (7, 63). Electron “leak” function, and the recently identified positive and negative roles
mainly occurs at ETC complex I and III, when elections of NOX isoforms in ␤-cell function (129), NOX dysregulation
derived from NADH or FADH2 that have escaped normal in T1DM and T2DM under conditions of proinflammatory
processing by the ETC react with molecular oxygen to form cytokines (such as IL-1␤, TNF-␣, and INF-␥) and glucose
O2·⫺, that is rapidly converted to H2O2 and subsequently excess can result in cellular dysfunction and impairment of
detoxified by CAT or GPx (see above), which essentially insulin secretion (120).
nullifies the initial toxicity from electron leak. However, this The availability of the NOX substrate NADPH, which is
flux may be disrupted in the ␤-cells, as they express relatively generally involved in biosynthetic pathways, for example fatty
low levels of CAT and Gpx (103), around 5% of that reported acid synthesis, and in the regeneration of reduced glutathione,
for liver cells (161). In addition, ␤-cells may also be chroni- is central to NOX activity. NADPH can be produced in the
cally exposed to glucose overload in the diabetic milieu, mitochondria by specific enzymes, but the vast majority of
leading to elevated glycolytic flux and glucose oxidation, cellular production comes from the pentose phosphate pathway
elevated tricarboxylic acid (TCA) cycle activity, and increased (PPP). In essence, generation of NADPH in high-glucose
Ca2⫹ oscillations. Together, these metabolic reactions increase conditions can be considered as a protective function, as the

AJP-Cell Physiol • doi:10.1152/ajpcell.00141.2019 • www.ajpcell.org


Downloaded from journals.physiology.org/journal/ajpcell (189.239.040.188) on May 14, 2022.
C422 OXIDATIVE STRESS, CELL METABOLISM AND FUNCTION

PPP shuttles excess glucose away from energy- and ROS- or smoked meats (96). With the ability to generate O2·⫺, other
generating processes (i.e., glycolysis and oxidative phosphor- ROS, and nitrosamines, cytochrome P450 enzymes have fur-
ylation) into biosynthetic pathways, while NADPH is also ther potential to generate highly reactive RNS (71, 75) and
required for converting oxidized glutathione into the reduced have been reported to increase DNA damage in BRIN-BD11
state so that it can nullify additional ROS species. However, ␤-cells in vitro (96). Nitric oxide can regulate insulin secretion
excessive NADPH generated via nutrient overload may also by enhancing insulin granule exocytosis through S-nitrosyla-
lead to elevated O2·⫺ production by promoting NOX activity. tion of syntaxin 4, a protein regulating vesicle-plasma mem-
Furthermore, it is understood that, following exposure of brane docking (173). However, these regulatory aspects are
␤-cells to glucose, there is a larger increase in the NADPH/ generally mediated by low to medium ROS/RNS levels, and it
NADP ratio compared with the NADH/NAD ratio (63), and has been reported in the INS-1 ␤-cell line that H2O2 in the
consequently NOX is activated in response to glucose stimu- range of 1– 4 ␮M can promote insulin release, while levels
lation (114). NOX also functions as a useful mechanism to above or below this range impede insulin exocytosis (63, 135).
regenerate NADP from NADPH, thus allowing further metab- ␤-Cell signaling pathways are exquisitely designed to link
olism of glucose via the PPP, albeit producing O2·⫺ in the nutrient-sensing capabilities to insulin secretion, the primary
process. This is particularly important as ␤-cells possess a function of the cell. This is achieved by coupling rapid uptake
relatively low level of lactate dehydrogenase (LDH) and con- and oxidative degradation of glucose to intracellular ATP
sequently are limited in their ability to regenerate NAD⫹ from production, which, via interaction with ATP sensitive K⫹
NADH through lactate production to promote glycolytic flux. channels, leads to plasma membrane depolarization, Ca2⫹
Key experiments have shown that inhibition of NOX (77, 115) influx, and appropriate insulin exocytosis (119). Elevated glu-
and other extramitochondrial oxidoreductase enzymes (63) cose entering ␤-cells is mainly oxidized to CO2 through gly-
impaired insulin release, as NOX is required for optimal GSIS colysis and TCA activity (53, 150), but it can also enhance
via O2·⫺ production (115). The regulation of NOX in ␤-cell anaplerosis and thus biosynthetic pathways, through formation
function is complex and is possibly dependent on the genera- of ␣-ketoglutarate, oxaloacetate, and citrate required for pro-
tion of other intracellular ROS species, such as H2O2 (115), as tein (150) and lipid synthesis (116), but, as described above, it
well as the level of glucose. Priming of the antioxidant defense increases the probability of electron leak and ROS generation.
system by low/initial ROS levels may be important for subse-
quent elevations of ROS generated by the enhancement of Other ROS Species and Their Sources
metabolic activity. Interestingly, using fasting and fed-state
animals, it was reported that increased NOX activity and ROS Given that O2·⫺ is the main source of cellular ROS species,
generation prevented hypoglycemia in fasting conditions via a it is important to also consider that it is a central regulator in
reduction in insulin output (117). Conversely, in high-glucose the generation of other ROS/RNS molecules. As illustrated
or leucine-stimulated conditions, the increased production of above, O2·⫺ can be converted to H2O2 through the action of
NADPH was diverted from NOX activity and into antioxidant SOD, and H2O2 is then detoxified by catalase. However, in
defense, which mitigated excess ROS production and was circumstances where O2·⫺-to-H2O2 fluxes are elevated but the
important for continued insulin secretion (117). Therefore, it subsequent H2O2 detoxifying activity of catalase is low [which
would appear that NOX activity, NADPH, O2·⫺, and their is the case for ␤-cells (103)], then both species can react
related fluxes based on nutrient or proinflammatory cytokine together to form the highly reactive hydroxyl radical (HO·) and
stimulation are intimately involved in ␤-cell function and fate. hydroxide (52, 122, 149). Importantly, this Fenton chemistry
Finally, ␤-cell O2·⫺ can also be produced in other cell reaction will only occur in the presence of metal ions, partic-
compartments by additional oxidase enzymes, including in the ularly iron (Fe2⫹ and Fe3⫹), and the probability of it occurring
cytosol by xanthine oxidase (XOD) (179), and in the smooth rises with increased intracellular metal ion concentrations (7,
endoplasmic reticulum (SER) by cytochrome P450 enzymes 163). This has implications for disorders including hemochro-
(23, 126). A recent in vitro study demonstrated that intermittent matosis and diabetes, where iron overload is or can be present,
high glucose (11.1 to 25 mM rotating every 24 h), significantly respectively (157), and the risk is intensified within glucotoxic,
increased XOD activity and O2·⫺ in the INS-1 ␤-cell line lipotoxic, or glucolipotoxic environments due to high meta-
(179). This also led to decreased cell viability and an accumu- bolic activity. In addition, O2·⫺ can react with ·NO to form a
lation of cells in the G0/G1 cell cycle stage. It was indicated dangerous RNS species, peroxynitrite (ONOO⫺) (67, 122).
that large fluctuations in glucose concentration were more Both HO· and ONOO⫺ are highly reactive species that can
detrimental to ␤-cell function than chronically elevated levels. rapidly promote significant cellular damage in rat and human
This may have implications for those with uncontrolled blood islets in vitro (29, 37, 67), and the latter was reported to be
glucose, although the functionality of the GSIS response in elevated in the islets of NOD mice (139). However, while HO·
these treated cells was not explored. Cytochrome P450 en- and ONOO⫺ may play a key role in ␤-cell oxidative stress (29,
zymes play a key role in detoxifying compounds in the SER 37, 67), HO· production is more relevant in response to cyto-
and are ubiquitously expressed in the liver but are also present kine insult, inflammation, and cellular dysfunction, as it was
in human islets (48, 96). Some SER enzymes, such as reported that ONOO⫺ was not generated by ␤-cells under
CYP2E1, have been demonstrated to be a risk factor for proinflammatory conditions (12, 67).
T2DM, and increased hepatic expression has been shown in ␤-Cell production of HO· can also occur via other mecha-
obesity (104, 171). The activity of these enzymes can directly nisms, including the reaction of ·NO or nitrite with H2O2
promote O2·⫺ and other ROS, but they can also be involved in (generated by high levels of SOD and reduced catalase, as
bioactivation of toxic compounds such as nitrosamines, that are outlined above) (67, 122). In this context, exposure of RINm5f
usually derived from poor nutritional sources such as processed ␤-cell lines to IL-1␤ alone induced ·NO production via the

AJP-Cell Physiol • doi:10.1152/ajpcell.00141.2019 • www.ajpcell.org


Downloaded from journals.physiology.org/journal/ajpcell (189.239.040.188) on May 14, 2022.
OXIDATIVE STRESS, CELL METABOLISM AND FUNCTION C423
expression of inducible nitric oxide synthase (iNOS), while the context of H2O2-induced impaired ␤-cell function. Utilizing
addition of TNF␣ and INF␥ increased the mitochondrial accu- INS-1E and RINm5f ␤-cell lines along with rat islets, Elsner et
mulation of H2O2. This proinflammatory combination led to al. (42) demonstrated that the peroxisome is a major site of
the production of HO·, which was highly toxic to the ␤-cells. H2O2 production in ␤-cells and that only saturated FFA with
However, this cytokine-mediated toxicity was abrogated by the carbon lengths greater than 14 were toxic to ␤-cells. Interest-
supply of catalase (67), demonstrating the importance of raised ingly, peroxisomal and cytosolic catalase overexpression pro-
H2O2 accumulation and reduced catalase activity in ␤-cell tected these cells from palmitate-induced toxicity by reducing
physiology. Additional work also revealed that ␤-cells respond the intracellular formation of H2O2. However, overexpression
to cytokine challenge by changing the expression status of of catalase in the mitochondria did not alter H2O2 levels and
RNS-generating enzymes such as the various isoforms of NOS. did not protect ␤-cells (42). Conversely, other research indi-
The constitutive form of NOS in ␤-cells, neuronal NOS cated that the production of H2O2 may be required for appro-
(nNOS), is mainly expressed within insulin vesicles (66, 94) priate intracellular signaling to allow insulin release. It can
and catalyzes the conversion of L-arginine to ·NO and L-citrul- positively modulate mitochondrial Ca2⫹ flux (97), which is a
line, using molecular oxygen and NADPH as substrates (120). major regulator of insulin release in ␤-cells (79). Furthermore,
However, when challenged with cytokines, nNOS expression Morgan et al. (115) also reported that suppression of NOX in
decreased in ␤-cells as a compensatory mechanism to offset a conditions of reduced H2O2 production led to a decrease in
cytokine-mediated rise in iNOS activity (66). So in this sce- GSIS. Recent investigations have demonstrated that insulin can
nario, a RNS-generating enzyme played a protective role in the potentiate the toxicity of H2O2 insult in ␤-cells. Exposure of
cell by reducing one source of RNS in response to stress. MIN-6 and RINm5f to insulin (100 nM) alone increased cell
Conversely, while some studies have demonstrated that induc- death over 24, 48, and 72 h (147), while exposure to a
ible production of ·NO is detrimental to insulin secretion (27) combination of insulin and H2O2 (70 ␮M) resulted in further
and ␤-cell proliferation (138), it is apparent that endogenous cell death over and above exposure to the same concentration
·
NO production is a physiological regulator of insulin secretion of H2O2 alone (147). Additional studies in rodent and human
in ␤-cells at lower concentrations (153). The production of ·NO islets confirmed that the cytotoxic response occurred concom-
by nNOS is important for liberation of glucokinase from itantly with markers of ER stress and alterations in BCL
insulin-secretory granules to the cytosol via nitrosylation proteins (15). These findings have stimulated interest with
(144). Gucokinase is a key rate-determining enzyme and as regard to whether or not the early overproduction and secretion
such is required for appropriate glycolytic flux and for gener- of insulin in obese and insulin-resistant humans may harm
ating a high ATP/ADP ratio, needed for insulin release. Inter- ␤-cell function (4). Taken together, there appears to be a
estingly, ·NO donors such as hydroxylamine and 3-morpholi- delicate balance between ROS/RNS production and detoxifi-
nosydnonimine also promoted insulin release in INS-1 cells. It cation that is critical for ␤-cell physiology. Further descriptions
was observed that intracellular ·NO levels were increased in of ROS and their relevant chemistry can be found in a recent
response to glucose and Ca2⫹ influx in INS-1 ␤-cells and rat review published in the Biochemical Journal (120).
islets, while scavenging of ·NO blocked insulin secretion (153).
Thus, low levels are involved in physiological signaling, while SOD, CAT, and GPx
higher levels are detrimental to ␤-cell function. This is due to
the high reactivity of ·NO and its promotion of nitrosative Antioxidants are required to attenuate and/or neutralize the
stress, which can target heme-containing proteins such as those deleterious effects of reactive substances, and can be grouped
in the ETC. In fact, ·NO can compete with O2 for binding to as enzymatic and nonenzymatic factors. Superoxide dismutase
ETC complex IV, changing the ⌬␺, ⌬pH, and Ca2⫹ retention (SOD, EC 1.15.1.1), catalase (CAT, EC 1.11.1.6), and gluta-
(54), while it can also induce S-nitrosylation of ETC com- thione-peroxidases (GPx, EC 1.11.1.9) are the main enzymatic
plexes I, III, and IV and the Fo/F1 ATPase (38, 54). These antioxidants and have been investigated for the treatment of
covalent modifications alter protein structure and thus function, diseases and catabolic circumstances resulting from oxidative
leading to impaired ATP generation and optimum insulin damage and/or oxidative stress (1).
release. As previously described, high concentrations and/or inade-
Finally, H2O2 is a nonradical ROS and is chemically more quate removal of reactive products, especially O2·⫺ results in
stable in comparison to other ROS species (134). H2O2 is oxidative stress, which has been a cause and/or a consequence
cytotoxic and elicits toxic effects through protein modification of most pathological conditions, such as cardiovascular dis-
via its interaction with protein thiol groups (121). While H2O2 eases, cancer, atherosclerosis, hypertension, and type 1 and 2
does not promote changes in DNA structure directly, the diabetes (40, 46). For instance, some important RNS signal
H2O2-induced production of HO·, as indicated above (134), can transduction molecules, such as ·NO, can be rapidly removed
lead to DNA damage and cell death. The formation of H2O2 by reacting with O2·⫺, resulting in the production of ONOO⫺,
can occur in various subcellular compartments mainly due to a compound with powerful oxidant properties (see above). This
the distribution of different SOD isoforms and the subcellular reaction highlights the importance of all compartmentalized
generation of O2·⫺ (mitochondria and cytosol). However, it can SOD isoforms as the first line of defense against oxidative
also be produced by other biochemical reactions including the stress.
degradation of FFA by ␤-oxidation in peroxisomes (134). Pharmacological treatments with SOD (39) and/or SOD
Various peroxisomal enzymes have the capacity to generate mimetics [e.g., Mn(II)(pyane)Cl2] (155) was reported to reduce
H2O2, including acyl CoA-oxidase, D-aspartate oxidase, L-a- liver and hippocampus oxidative stress, respectively, in dia-
hydroxy oxidase, and XOD (134), but it is possibly those betic animals. Studies also report antioxidant effects using
involved in FFA metabolism that are more pertinent in the chemically modified SOD (e.g., carboxymethylcellulose-SOD,

AJP-Cell Physiol • doi:10.1152/ajpcell.00141.2019 • www.ajpcell.org


Downloaded from journals.physiology.org/journal/ajpcell (189.239.040.188) on May 14, 2022.
C424 OXIDATIVE STRESS, CELL METABOLISM AND FUNCTION

and polymethyl vinyl ether-co-maleic anhydride-SOD) in The phenotype exhibited by Gx1⫺/⫺ mice is considered closely
streptozotocin (STZ)-induced diabetic rats (108). The defi- associated with cell dysfunction described for type 1 diabetes
ciency of some trace minerals such as zinc has also been (102, 170).
implicated in the effectiveness of Cu-Zn-SOD and therefore The activity of GPx is dependent on the availability of
associated with reduced insulin secretion and action (100). For micronutrients such as selenium (Se). Se concentration in the
instance, long-term low zinc intake from drinking groundwater bloodstream has been used to assess whole body Se status and
was reported to be associated with an increased risk of devel- as an indirect antioxidant indicator of GPx activity. Many in
oping type 1 diabetes (6, 156). In obese animal models, such as vivo and in vitro studies demonstrated the potential of seleno-
mutant db/db mice and mice subjected to a high-fat diet (HFD), proteins and GPx-related antioxidant effects induced by sele-
however, zinc supplementation improved glucose homeostasis nium and therefore led to the idea that an increased Se intake
by restoration of ␤-cell function (25, 151). through the diet or supplementation would attenuate the oxi-
CAT is an antioxidant enzyme present in almost all aerobic dative stress observed in diabetes (143). However, later stud-
organisms and is considered the detoxification-completing step ies, including epidemiological studies, observed that both ex-
initiated by SOD. It is primarily located in the peroxisomes and cessive or low selenium intake is associated with increased risk
absent in the mitochondria of almost all tissues (except in the of diabetes, following a U-shaped curve (76, 128, 142, 143).
heart) (21, 140). The main function of CAT is to catalyze the This is particularly important for well-nourished populations,
degradation/reduction of H2O2 to H2O and molecular oxygen. where Se deficiency is less likely to occur (76, 143), and
In addition, CAT reacts with H⫹ donors, such as methanol, therefore a nutritional assessment associated with the determi-
ethanol, formic acid, or phenols, with peroxidase activity. nation of Se concentration in plasma of diabetic patients is
Specific mutations/polymorphisms in CAT gene may result in recommended before considering increments in Se intake.
catalase deficiency (also known as acatalasemia), as reported in
Japanese, Swiss, and Hungarian individuals (59, 60, 158). The Gene Expression Mechanisms Responsible for ROS Effects
deficiency or mutation of CAT has been associated with low on Insulin Secretion
H2O2 removal and, therefore, oxidative stress leading to vari-
ous diseases and abnormalities, such atherosclerosis, hyperlip- It is clear that ROS/RNS act as signaling molecules and that
idemia, gangrene, hypertension, neurodegeneration, and diabe- elevated levels negatively affect ␤-cell function including in-
tes (60). At the ␤-cell level, low CAT level results in the sulin secretion (Fig. 1). However, the precise mechanisms by
inability to protect the pancreatic ␤-cells from toxic quantities which ROS elicit these effects are complex. Due to their highly
of H2O2; however, this is more likely to be observed when reactive nature, ROS can alter the native conformation of
␤-cells are stimulated with glucose. Furthermore, acatalasemic proteins by chemical modification like nitrosylation, carbony-
mice treated with alloxan are more prone to exhibit low insulin lation, and peroxidation among others, and this leads to im-
level due to the decline in the number of viable ␤-cells paired function. Some of the most common targets include
(compared with normal-CAT animals) (159). Although con- proteins associated with membrane ion transport, metabolic
ventional antioxidant treatments for low CAT activity are enzymes, and proteins regulating gene expression and signal
available, there is a lack of scientific evidence evaluating their transduction (53, 122). For example, H2O2 modulates proin-
effect. In addition, conventional antioxidant treatments for flammatory NF-␬B signaling and regulates its translocation
acatalasemic-induced type 1 or 2 diabetes may increase ROS into the nucleus. However, the effects are dependent on the
synthesis through endomembrane signaling cascades, commu- concentration of H2O2 and the inflammatory conditions. For
nication networks, and metabolic regulatory complexes. A instance, H2O2 alone does not promote nuclear translocation of
recent alternative has been studied by using a cell-penetrating NF-␬B but enhances this process in the presence of TNF (36).
derivative of the peroxisomal enzyme catalase (also called Conversely, when concentrations were increased, translocation
CAT-SKL). This highly specific supplementation has shown a was inhibited (36). Other studies have reported that oxidized
significant reduction in oxidative stress associated with dia- NF-␬B had a significantly reduced ability to bind DNA and
betic retinopathy in in vitro and in vivo animal models (55). mediate gene transcription, although this was restored with
H2O2 conversion to H2O and molecular oxygen can also antioxidant enzymes (83). More recent work has demonstrated
occur through GPx (selenium-dependent glutathione peroxi- that NOX-generated ROS was critical for redox balance and
dase). There are at least eight types of GPx, namely GPx1– 8, reduced the LPS-induced interaction of NF-␬B with DNA,
however, the most well described and abundant in cells is rather than altering the nuclear translocation (68). The ability
GPx1. This antioxidant enzyme is mainly located in mamma- of NF-␬B to interact with DNA is influenced by the redox
lian cell mitochondria, and to a lesser extent in the cytosol. status of cysteine moieties located within various subunits,
GPx also detoxifies the cell from organic and inorganic per- including Cys62 of p50 and Cys38 of p65 (56). Thiol groups
oxides and therefore provides protection against a variety of and cysteine residues are sensitive to redox homeostasis and
oxidative stressors (76). In Gpx1⫺/⫺ mice, Wang et al. (170) are key targets for ROS (73), which alters subsequent activity.
observed a significant reduction in ␤-cell mass and GSIS, One of the classical examples of ROS-mediated negative
allied with hyperinsulinemia, mild hyperglycemia, and im- effects on ␤-cell function and insulin secretion was a report of
paired ATP production. In another study, Gpx1⫺/⫺ mice sub- ROS impacting insulin gene expression via major redox-sen-
jected to HFD for 12 wk exhibited chronic and systemic sitive transcription factors, pancreatic and duodenal homeobox
oxidative stress associated with hyperglycemia. Although no 1 (PDX1), and V-Maf avian musculoaponeurotic fibrosarcoma
alteration in insulin sensitivity was observed, chronic oxidative oncogene homolog A (MafA) (24). Both are targets for ROS
stress induced by ROS dramatically reduced ␤-cell mass and (85, 121), and exposure of rat islets to H2O2 reduced the
insulin secretion associated with Pdx1 gene expression (112). binding interaction of PDX-1 with DNA, leading to decreased

AJP-Cell Physiol • doi:10.1152/ajpcell.00141.2019 • www.ajpcell.org


Downloaded from journals.physiology.org/journal/ajpcell (189.239.040.188) on May 14, 2022.
OXIDATIVE STRESS, CELL METABOLISM AND FUNCTION C425

Fig. 1. Interplay between nutrient metabolism and reactive oxygen species (ROS) generation in regulating ␤-cell function. Exposure to high nutrient load
including glucose (Gluc) and free fatty acids (FFA) leads to enhanced mitochondrial metabolism and possibly increased ROS production in ␤-cells. If not
neutralized by the antioxidant arsenal, elevated ROS levels may alter electron transport efficiency, decreasing ATP production and raising the mitochondrial
membrane potential (MMP), which leads to electron slippage from normal transport processes. The reduced ability of ␤-cells to increase or maintain ATP levels
in response to nutrient stimulation may lead to impaired insulin secretion through decreased action on ATP-sensitive K⫹ channels. Elevated ROS generation
through other mechanisms, such as the proinflammatory NADPH oxidase (NOX) activity, may also impair insulin gene transcription and biosynthesis,
contributing to reduced insulin output. However, increased ROS may activate the Keap1-Nrf2 antioxidant pathway by enhancing the cytoplasmic to nuclear
translocation of Nrf2. Gln, glutamine; Arg, arginine, Glu, glutamine; iNOS, inducible nitric oxide synthase; nNOS, neuronal nitric oxide synthase; GCK,
glucokinase; Cat, catalase; Gpx, glutathione peroxidase; ARE, antioxidant response elements; SOD, superoxide dismutase.

insulin gene expression (85). For PDX-1, it appears that the localization of MafA and prevented its positive influence on
negative effects of ROS are mediated through the c-Jun NH2- insulin gene expression (169). Work in animal models demon-
terminal kinase (JNK) signaling and the forkhead box protein strated that overexpression of antioxidants such as GPx main-
O1 (FOXO1) transcription factor (53, 84). Inhibition of the tained nuclear expression of MafA and ␤-cell function (65). It was
JNK pathway protected ␤-cells from ROS, while activation proposed that in hyperglycemic conditions the cytoplasmic trans-
reduced insulin gene expression (84, 85). Exposure of HIT-T15 location of MafA is an early indicator of ␤-cell demise (65, 169).
␤-cells to H2O2 for 48 h led the cytoplasmic-nuclear translo- Therefore, it appears that redox status can affect the opposing
cation of FOXO1, while PDX-1 was expressed mainly in the subcellular localization of key insulin gene transcription factors
cytoplasm. In addition, JNK activation also induced nuclear and their potential regulatory proteins.
translocation of FOXO1, whereas inhibition of FOXO1 expres-
sion with siRNA maintained nuclear expression of PDX-1 and Novel ␤-Cell Cell-Antioxidant Defenses in Diabetes: the
importantly insulin secretion (86). A similar observation was Nrf2-Keap1 Pathway
reported with respect to the transcription factor MafA, which
regulates insulin gene transcription along with ␤-cell development Redox homeostasis/balance is essential for cell homeostasis
and proliferation (125, 169). H2O2 also promoted cytoplasmic (30, 78, 98, 113, 137), and in the case of pancreatic ␤-cells it

AJP-Cell Physiol • doi:10.1152/ajpcell.00141.2019 • www.ajpcell.org


Downloaded from journals.physiology.org/journal/ajpcell (189.239.040.188) on May 14, 2022.
C426 OXIDATIVE STRESS, CELL METABOLISM AND FUNCTION

is very important to minimize the deleterious effects of excess contribute to the resistance and survival of supercells and
nutrients or inflammatory factors on ␤-cell function, i.e., insu- therefore the evolution of cancer and chemoresistance (69).
lin secretion and cell death (see above). In normal or elevated Furthermore, the balance of T helper cell (Th)1 and Th2
nutrient/immune stress circumstances, redox component levels immune responses are critical for immunity, and studies have
are regulated at the transcriptional level of the cell. The shown that both responses can be regulated by the Nrf2
Nrf2/Keap1 pathway regulates the expression of over 100 downstream pathway in different ways. For instance, Rockwell
genes and functions related to oxidative stress and cell sur- et al. (145) and Kikuchi et al. (87) reported that Nrf2 promotes
vival, including the above mentioned enzymatic and non- Th2 cytokine production via Th1 response suppression in mice
enzymatic antioxidants, growth factors and transcription fac- exposed to food preservatives (i.e., allergy animal model), and
tors related to inflammation (Fig. 1) (34). Indeed the role of the pulmonary fibrosis, respectively. However, in diabetic condi-
Nrf2/Keap1 pathway in mediating redox balance in ␤-cells has tions (1, 2), the demise of pancreatic ␤-cells appears to occur
been further explored, with dysregulation contributing to dia- through the Th1 cytokine cascade and results in cytotoxic
betic pathologies (34). In basal conditions, Keap1 marks the quantities of ROS/RNS (93, 131). Sireesh et al. (152) observed
Nrf2 transcription factor for ubiquitination in the cytoplasm. a positive correlation between plasma Nrf2 and Th2 cytokine
After oxidative insult, redox-sensitive changes in Keap1 cys- (IL-4, IL-13) levels, and a negative correlation with Th1
teine moieties lead to the dissociation of the Keap1-Nrf2 cytokine (IFN-␥, TNF) levels in early diagnosed type 2 diabe-
dimer, allowing the entry of Nrf2 into the nucleus, where tes patients. These findings support other type 1 (72, 93, 127,
various genes are upregulated, including those involved in 131) and type 2 (34, 105) diabetes studies, where reduced
antioxidant defenses such as ␥-glutamylcysteine synthetase levels of Nrf2 favors Th1 responses and oxidative stress,
(GCLC), glutathione S-transferase-A2 (GSTA2), NADPH qui- potentially contributing to the progression of the disease.
none oxidoreductase (NQO-1), superoxide dismutase (SOD1),
and heme oxygenase-1 (HO-1)(137). Murine studies have INSULIN RESISTANCE
reported that knockout and suppression of Nrf2 led to increased ROS in the Periphery and Their Role in Insulin Resistance
intracellular ROS and aggravated ␤-cell damage in islets and
␤-cells lines (174). Conversely, overexpression of Nrf2 re- Insulin resistance is the pathological condition in which periph-
duced DNA adduct formation and reestablished insulin secre- eral tissues (e.g., liver, skeletal muscle, and adipose tissue) fail to
tion (174). This pathway also reduced inflammation associated respond to normal levels of insulin. As a result, pancreatic ␤-cells
with NF-␬B activation (154), which indicated effects beyond have to produce and secrete higher amounts of insulin to com-
redox balance. However, the precise effects of this pathway on pensate. This poses a significant biosynthetic and energy burden
insulin-secretory machinery and insulin release and homeosta- on the ␤-cells. When ␤-cells fail to maintain high insulin produc-
sis are not fully understood, as in Nrf2 knockout mice, de- tion in response to greater metabolic demand, T2DM results
creased blood glucose, lower body fat and body weight along (120). There is a large body of evidence connecting hyperglyce-
with enhanced insulin signaling was observed (136, 178). mia and many of the major complications of diabetes including
Several studies have reported changes in Nrf2 expression in nephropathy, retinopathy, neuropathy, and macro- and microvas-
diabetic individuals, and the direction of these changes appears cular damage (13, 120). In uncontrolled diabetes, decreased levels
to be related to the presence of diabetic complications and of the antioxidants vitamin E and ␣-lipoic acid (LA) have been
response to oxidative stress conditions, including upregulation reported (130). There is also evidence of a deficiency in erythro-
in diabetic nephropathy and retinopathy (80, 180) and down- cyte catalase, an enzyme responsible for the removal of H2O2, that
regulation in the diabetic myocardium (160). Although the role can occur in diabetes (60, 61). In vivo studies demonstrate that
played by Nrf2/Keap1 in mediating oxidative stress in diabetes oxidative stress due to hyperglycemia occurs before late compli-
remains to be fully understood, the pathway has been consid- cations are clinically evident (123). The stress-activated signaling
ered a target for pharmacological and nutritional therapeutics. pathways indicated above, including NF-␬B, p38 MAPK, JNK/
For example, the drug dimethyl fumarate (BG-12) is an acti- stress-activated protein kinases (SAPK), advanced glycosylation
vator of Nrf2 downstream pathways, leading to cytoprotective, end-products (AGE)/receptor for AGE (RAGE), and PKC may all
anti-inflammatory, and antioxidant effects and is already a be activated by oxidative stress. ROS generation is critical for
target for the treatment of multiple sclerosis (57). The nutri- mediating hyperglycemia-induced cellular damage (124). In bo-
tional approach also shed light in some specific nutrient acti- vine endothelial cells, exposure to hyperglycemia initially in-
vators of Nrf2, including vitamin D, zinc, and bioactive com- creased the production of intracellular ROS and activated NF-␬B.
pounds found in foods and herbs (e.g., curcumin, sulforaphane, Subsequently, PKC activity, AGE, and sorbitol levels increased. It
cinnamic aldehyde, resveratrol, magnesium lithospermate B, is likely that oxidative stress occurs early in the sequence of events
catechins, pterostilbene) (5, 34, 81). Furthermore, some amino induced by high glucose, followed by activation of other pathways
acids such as glutamine and arginine exhibit Nrf2-activating that lead to cellular dysfunction and damage (120, 123).
properties in proliferating cells of the intestinal crypt of healthy Studies in human populations have described a strong link
animals (167) and in left ventricular heart tissue of alloxan- between insulin resistance and elevated oxidative stress (43,
induced hyperglycemic rats (141), respectively. Interestingly, 58, 132, 133, 165). Using a euglycemic-hyperinsulinemia
arginine and glutamine are also sources of glutamate, the clamp approach, (considered to be the best method to estimate
supply of which can be rate limiting for the synthesis of even small variations in insulin sensitivity (88), in a population
glutathione (GSH) through GCLC in oxidative stress circum- of obese and nonobese men, Urakawa et al. (165), observed a
stances (30, 31, 146). The Nrf2/Keap1/ARE pathway is also a positive association between insulin resistance and circulating
target in cancer/tumor cells and autoimmune diseases, includ- levels of 8-epi-PGF2␣, a marker of lipid peroxidation. Differ-
ing T1DM. The activation of Nrf2 downstream cascades may ent models to assess insulin resistance, such as the homeostasis

AJP-Cell Physiol • doi:10.1152/ajpcell.00141.2019 • www.ajpcell.org


Downloaded from journals.physiology.org/journal/ajpcell (189.239.040.188) on May 14, 2022.
OXIDATIVE STRESS, CELL METABOLISM AND FUNCTION C427
model assessment of insulin resistance (HOMA-IR) have also Thus, oxidative stress may indeed play a causal role in the
been reported to positively associate with oxidative stress development of insulin resistance. Houstis et al. (74) used two
levels in various populations (89, 110, 132, 133). Additionally, models to induce insulin resistance in adipocytes, TNF and
genes involved in providing cellular protection against oxida- dexamethasone treatment. Importantly, in both models, ele-
tive stress are often found to be dysregulated in both insulin- vated ROS production was detected before any insulin insen-
resistant and T2DM patients (2, 14, 22). For instance, the sitivity could be observed, suggesting that exacerbated oxida-
expression levels of Mn-SOD in skeletal muscle tissue have tive stress is upstream of insulin signaling defects. In the same
been reported to be significantly decreased in obese patients study, insulin resistance was successfully ameliorated to vary-
and also in pregnant women that were overweight or were ing degrees using various distinct approaches designed to
clinically diagnosed with gestational diabetes (11, 44). In reduce cellular ROS levels, highlighting that insulin resistance
support of this view, overexpression of Mn-SOD ameliorated can be attenuated by suppression of redox stress. Not surpris-
insulin sensitivity in a rat model of HFD-induced skeletal ing, perhaps, is the ever-increasing list of publications provid-
muscle insulin resistance (8). Conversely, downregulation of ing direct and indirect evidence that a reduction in intracellular
redox control enzymes or antioxidant protein-associated tran- ROS levels, either through genetic manipulations or through
scriptional factors such as Nrf2, has been reported to contribute the use of antioxidants, can alleviate insulin resistance and
to oxidative stress-induced insulin resistance (160). glucose intolerance (3, 20, 70, 95, 106).
In this scenario, ectopic lipid accumulation and mitochon- Mitochondrial electron transport overload due to excess
drial dysfunction, particularly in the skeletal muscle tissue, are dietary nutrient metabolism appears to be central to the mech-
important physiological hallmarks of insulin-resistant subjects. anism leading to elevated ROS generation, as described above
They have been reported to be paralleled and also preceded by (1, 162). Incomplete fatty acid metabolism is associated with
increased ROS production in the latter sites (9, 109) (Fig. 2). accumulation of ROS that can directly impair insulin receptor

↑↑ Pro-inflammatory ↑ Impaired glucose disposal


Insulin
↑ Hyperinsulinemia
cytokines Gluc
e.g.TNF-α 7 Gluc
IL-6 Insulin Gluc 1 ↑ Ectopic lipid
G LU accumulation
IL-1β Receptor T4
Adipokines
1

ine
y tok tors Tyrosine
C ep
c phosphorylation
Re
IRS ↓ ↓ GLUT 4 translocation
2
↑ Fatty acid
↓ Glucose
↑ IRS-1 serine phosphorylation transport into
uptake
PI3K ↓ mitochondria
↑ JKK, p38, IKKβ, NFκB, PKC
5 T
PDK1 ↓ G
3
↑↑ Glut 4 vesicle
↑ Beta-oxidation
↑↑ ETC ↑↑ ETC
GSK3 AKT ↓
4
6 Mitochondrial
↑↑ dysfunction
↓ Glycogen synthesis and storage M
Metabolic
by
by-products

↓ Represent processes reduced in insulin resistance


↑ Represent processes exacerbated in insulin resistance
Skeletal muscles
Fig. 2. Role of reactive oxygen species (ROS) in lipid-induced skeletal muscle insulin resistance. Ectopic lipid accumulation leads to increased fatty acid transport
into the sarcoplasm and mitochondria of skeletal muscle cells. Excess fatty acids are metabolized through ␤-oxidation, resulting in mitochondrial electron
transport overload and increased formation of metabolic by-products and ROS. Stress response kinases (JNK, p38, PKC) are activated by ROS and can directly
impair insulin receptor signaling through serine phosphorylation of insulin receptor substrate 1 (IRS-1). This, in turn, inhibits tyrosine phosphorylation of IRS-1
in response to insulin binding, subsequently blocking downstream insulin signaling through PI3K/PDK1/Akt and, consequently, GLUT-4 translocation as well
as glycogen synthase activation. As a result, there is a decrease in insulin-stimulated glucose transport and glycogen synthesis, all together leading to impaired
glucose disposal and insulin hypersecretion as a compensatory mechanism. Red arrows pointing down represent processes reduced in insulin resistance; blue
arrows pointing up represent processes exacerbated in insulin resistance. Akt, protein kinase B; ETC, electron transport chain; Gluc, glucose; GLUT-4, glucose
transporter type 4; GSK-3, glycogen synthase kinase-3; IKK-␤, inhibitor of NF-␬B kinase-␤; IRS-1, insulin receptor substrate-1; JNK, c-Jun NH2-terminal
kinase; NF-␬B, nuclear factor-␬B; PDK1, phosphoinositide-dependent kinase-1; PI3K, phosphoinositide 3-kinase; PKC, protein kinase C; RNS, reactive nitrogen
species; ROS, reactive oxygen species.

AJP-Cell Physiol • doi:10.1152/ajpcell.00141.2019 • www.ajpcell.org


Downloaded from journals.physiology.org/journal/ajpcell (189.239.040.188) on May 14, 2022.
C428 OXIDATIVE STRESS, CELL METABOLISM AND FUNCTION

signaling through activation of several of the protein kinases domized controlled trials involving 283 participants and supple-
and transcription factors that control insulin sensitivity, such as mentation ranged from 4 wk to 12 mo (181).
JNK, p38 MAPK, I␬B kinase (IKK-␤), NF-␬B, and PKC
(177). In particular, JNK and p38 MAPK respectively can GSH and Mechanisms to Therapeutically Protect the Redox
directly and indirectly induce serine phosphorylation of insulin State
receptor substrate (IRS)-1/2 (35, 49, 50, 82, 118), an important Nonenzymatic antioxidants are a group of compounds that
negative feedback regulatory mechanism of insulin signaling can react directly or in conjunction with enzymatic antioxi-
(64), and widely reported to participate in cellular insulin dants against ROS and RNS. A variety of substances can be
resistance (41). Mechanistically, serine phosphorylation of classified as a nonenzymatic antioxidant, such as polyphenols,
IRS-1/2 induces their dissociation from the insulin receptor, ascorbic acid, vitamin A, ␣-lipoic acid, thioredoxin, melatonin,
ubiquitination, and proteasome degradation (26), ultimately coenzyme Q, ␤-carotenoids, and ␣-tocopherols (45). However,
promoting insulin resistance through reduced insulin signaling. the most important and most concentrated nonenzymatic anti-
Antioxidant supplementation has been frequently tested in oxidant ubiquitously distributed in eukaryotes is glutathione
human populations in various trials designed to evaluate their (␥-L-glutamyl-L-cysteinylglycine, GSH). GSH is composed of
efficacy to improve insulin sensitivity. Vitamins C, E, and three amino acid residues cysteine, glutamic acid, and glycine
␤-carotene have been tested either alone or in combination and and reacts with ROS in nonenzymatic reaction or as an electron
have been generally reported to improve insulin sensitivity donor in the peroxide reduction catalyzed by GPx (32, 47).
outcomes. A short-term 8-wk placebo-controlled trial reported Around 85–90% of GSH is found in the cytosol, and ~10 –15%
that a combination of vitamins C, E, and ␤-carotene supple- is located in organelles such as the mitochondria, nuclear
mentation resulted in a significant improvement in HOMA and matrix, and peroxisomes. Since GSH is a critical antioxidant,
a concomitant reduction in fasting insulin levels in young therapeutic approaches to increase systemic and/or tissue-
obese subjects (168). When tested alone, low doses (200 IU) of specific glutathione concentrations are of considerable interest,
vitamin E did not produce detectable improvements in fasting especially through supplementation studies involving glu-
insulin or glucose after 27 wk of supplementation in T2D tamine, GSH or N-acetylcysteine (NAC) (3, 18, 21, 33–35, 37,
patients (10). However, high doses of 800 IU resulted in 117, 136). For instance, in vitro and in vivo studies have
notable improvements in fasting plasma glucose and insulin demonstrated a reciprocal relationship between low glutamine
levels in overweight individuals at 3 mo of treatments, which availability and GSH in ␤-cells, skeletal muscle, and liver
was not sustained at 6 mo after the beginning of the supple- tissues, with a concomitant rise in oxidative stress (28, 111,
mentation regimen (107). Vitamin C when supplemented in 172). Importantly, a recent in vitro study by the Newsholme
combination with metformin has been demonstrated to consti- laboratory has shown that glutamine deprivation leads to rapid
tute a superior therapy in terms of glycemic control improve- induction of oxidative and endoplasmic reticulum stress, ulti-
ments compared with metformin alone (33). ␤-Carotene when mately resulting in marked ␤-cell dysfunction (18). However,
supplied in the form of a fruit and vegetable juice concentrate few studies in humans have demonstrated improvement in the
to overweight children attenuated insulin resistance, which was whole body and ␤-cell oxidative stress or glucose homeostasis.
also associated with a detectable increase in plasma levels of
␤-carotene (17). Omega 3 fatty acids also resulted in positive CONCLUDING REMARKS
outcomes when tested for their potential to ameliorate insulin This review has evaluated several metabolic cell signaling
resistance. A 90-day placebo-controlled trial demonstrated that mechanisms emphasizing the control of the pathways of mito-
1,800 mg of a combination of ecosapentaenoic acid and doco- chondrial ROS generation, NADPH-oxidase-dependent ROS
sahexaenoic acid daily improved several glycemic control generation, and the acute and chronic roles of antioxidants in
markers in a heterogeneous population of T2D patients with reducing oxidative stress. The modulation of various steps of
5–10 yr of disease diagnostic (164). Indeed, a recent meta- ROS generation and subsequent signaling that are induced by
analysis pooling data from 17 studies with a total number of hyperglycemia could be potential therapeutical targets in dia-
672 participants revealed that short-term fish oil supplementa- betes, e.g., attenuating ROS generation, cytokine production,
tion is associated with improved insulin sensitivity in people and cellular dysfunction and death. It is known that, besides its
with various metabolic conditions (51). The proposed mecha- osmotic effects on cells, hyperglycemia activates mitochon-
nisms for the action of omega 3 fatty acids on insulin sensi- drial and NADPH-oxidase ROS production, which leads to
tivity likely involve their effects on mitochondrial bioenerget- oxidative stress in diabetes. Inflammatory reactions can be
ics and endoplasmic reticulum and have recently been expertly promoted by hyperglycemia ROS production and they may be
reviewed (99). Another antioxidant molecule, lycopene, a ca- aggravated by the action of AGEs, due to the interaction with
rotenoid pigment found in tomatoes, has recently been reported the responsible receptor (RAGE). ROS and AGE generation
in animal models to improve insulin sensitivity, although can lead to chronic low-grade inflammation that may contrib-
clinical evidence is still lacking to support its use in humans for ute to diabetic complications. Any attempt to control these
the specific purpose of controlling insulin resistance (175, processes could modulate some of the consequences of hyper-
176). Resveratrol, traditionally associated with improvements glycemia. NADPH-oxidase-specific and antioxidant levels
in cardiovascular outcomes, was recently assessed in two system- have been suggested as being the best targets to reduce hyper-
atic review and meta-analysis studies evaluating its effects on glycemia-induced oxidative stress, but more targeted therapies
glucose homeostasis parameters in T2D patients. Insulin resis- need to be found. In addition, future translational studies in
tance as assessed by HOMA was significantly ameliorated in T2D humans should reveal whether some nutrient-based antioxi-
patients receiving resveratrol; data were pooled from nine ran- dants can attenuate oxidative stress occurring in diabetes.

AJP-Cell Physiol • doi:10.1152/ajpcell.00141.2019 • www.ajpcell.org


Downloaded from journals.physiology.org/journal/ajpcell (189.239.040.188) on May 14, 2022.
OXIDATIVE STRESS, CELL METABOLISM AND FUNCTION C429
ACKNOWLEDGMENTS 14. Bruce CR, Carey AL, Hawley JA, Febbraio MA. Intramuscular heat
shock protein 72 and heme oxygenase-1 mRNA are reduced in patients
The authors thank Curtin University School of Pharmacy and Biomedical
with type 2 diabetes: evidence that insulin resistance is associated with a
Sciences, Curtin Health Innovation Research Institute, and Faculty of Health,
disturbed antioxidant defense mechanism. Diabetes 52: 2338 –2345,
Torrens University Australia, for provision of excellent research facilities and
2003. doi:10.2337/diabetes.52.9.2338.
support. Part of this review was written while P. Newsholme was on Academic
15. Bucris E, Beck A, Boura-Halfon S, Isaac R, Vinik Y, Rosenzweig T,
Program Study Leave at the Rowett Institute, University of Aberdeen, Scot-
land. Sampson SR, Zick Y. Prolonged insulin treatment sensitizes apoptosis
pathways in pancreatic ␤ cells. J Endocrinol 230: 291–307, 2016.
DISCLOSURES doi:10.1530/JOE-15-0505.
16. Cadenas E, Davies KJA. Mitochondrial free radical generation, oxida-
No conflicts of interest, financial or otherwise, are declared by the authors. tive stress, and aging. Free Radic Biol Med 29: 222–230, 2000. doi:10.
AUTHOR CONTRIBUTIONS 1016/S0891-5849(00)00317-8.
17. Canas JA, Damaso L, Altomare A, Killen K, Hossain J, Balagopal
K.N.K., R.C., and V.C. prepared figures; P.N., K.N.K., R.C., and V.C. PB. Insulin resistance and adiposity in relation to serum ␤-carotene
drafted manuscript; P.N., K.N.K., R.C., and V.C. edited and revised manu- levels. J Pediatr 161: 58 –64.e2, 2012. doi:10.1016/j.jpeds.2012.01.030.
script; P.N., K.N.K., R.C., and V.C. approved final version of manuscript. 18. Carlessi R, Rowlands J, Ellison G, Helena de Oliveira Alves H, News-
REFERENCES holme P, Mamotte C. Glutamine deprivation induces metabolic adaptations
associated with ␤-cell dysfunction and exacerbate lipotoxicity. Mol Cell
1. Affourtit C. Mitochondrial involvement in skeletal muscle insulin resis- Endocrinol 491: 110433, 2019. doi:10.1016/j.mce.2019.04.013.
tance: a case of imbalanced bioenergetics. Biochim Biophys Acta 1857: 19. Carlsson C, Borg LA, Welsh N. Sodium palmitate induces partial
1678 –1693, 2016. doi:10.1016/j.bbabio.2016.07.008. mitochondrial uncoupling and reactive oxygen species in rat pancreatic
2. Alibegovic AC, Sonne MP, Højbjerre L, Bork-Jensen J, Jacobsen S, islets in vitro. Endocrinology 140: 3422–3428, 1999. doi:10.1210/endo.
Nilsson E, Faerch K, Hiscock N, Mortensen B, Friedrichsen M, 140.8.6908.
Stallknecht B, Dela F, Vaag A. Insulin resistance induced by physical 20. Chen L, Na R, Gu M, Salmon AB, Liu Y, Liang H, Qi W, Van
inactivity is associated with multiple transcriptional changes in skeletal Remmen H, Richardson A, Ran Q. Reduction of mitochondrial H2O2
muscle in young men. Am J Physiol Endocrinol Metab 299: E752–E763, by overexpressing peroxiredoxin 3 improves glucose tolerance in mice.
2010. doi:10.1152/ajpendo.00590.2009. Aging Cell 7: 866 –878, 2008. doi:10.1111/j.1474-9726.2008.00432.x.
3. Anhê FF, Roy D, Pilon G, Dudonné S, Matamoros S, Varin TV, 21. Chen T, Jin X, Crawford BH, Cheng H, Saafir TB, Wagner MB,
Garofalo C, Moine Q, Desjardins Y, Levy E, Marette A. A polyphe- Yuan Z, Ding G. Cardioprotection from oxidative stress in the newborn
nol-rich cranberry extract protects from diet-induced obesity, insulin heart by activation of PPAR␥ is mediated by catalase. Free Radic Biol
resistance and intestinal inflammation in association with increased Med 53: 208 –215, 2012. doi:10.1016/j.freeradbiomed.2012.05.014.
Akkermansia spp. population in the gut microbiota of mice. Gut 64: 22. Cheng X, Siow RC, Mann GE. Impaired redox signaling and antioxi-
872–883, 2015. doi:10.1136/gutjnl-2014-307142. dant gene expression in endothelial cells in diabetes: a role for mito-
4. Aston-Mourney K, Proietto J, Morahan G, Andrikopoulos S. Too chondria and the nuclear factor-E2-related factor 2-Kelch-like ECH-
much of a good thing: why it is bad to stimulate the ␤-cell to secrete associated protein 1 defense pathway. Antioxid Redox Signal 14: 469 –
insulin. Diabetologia 51: 540 –545, 2008. doi:10.1007/s00125-008- 487, 2011. doi:10.1089/ars.2010.3283.
0930-2. 23. Clarke J, Flatt PR, Barnett CR. Cytochrome P450 1A-like proteins
5. Battino M, Giampieri F, Pistollato F, Sureda A, de Oliveira MR, expressed in the islets of Langerhans and altered pancreatic ␤-cell
Pittalà V, Fallarino F, Nabavi SF, Atanasov AG, Nabavi SM. Nrf2 as secretory responsiveness. Br J Pharmacol 121: 389 –394, 1997. doi:10.
regulator of innate immunity: a molecular Swiss army knife! Biotechnol 1038/sj.bjp.0701139.
Adv 36: 358 –370, 2018. doi:10.1016/j.biotechadv.2017.12.012. 24. Cnop M, Abdulkarim B, Bottu G, Cunha DA, Igoillo-Esteve M,
6. Benson VS, Vanleeuwen JA, Taylor J, Somers GS, McKinney PA, Masini M, Turatsinze JV, Griebel T, Villate O, Santin I, Bugliani
Van Til L. Type 1 diabetes mellitus and components in drinking water M, Ladriere L, Marselli L, McCarthy MI, Marchetti P, Sammeth
and diet: a population-based, case-control study in Prince Edward Island, M, Eizirik DL. RNA sequencing identifies dysregulation of the human
Canada. J Am Coll Nutr 29: 612–624, 2010. doi:10.1080/07315724.2010. pancreatic islet transcriptome by the saturated fatty acid palmitate.
10719900.
Diabetes 63: 1978 –1993, 2014. doi:10.2337/db13-1383.
7. Bhattacharya S. Reactive oxygen species and cellular defense system.
25. Cooper-Capetini V, de Vasconcelos DAA, Martins AR, Hirabara
In: Free Radicals in Human Health and Disease, edited by Rani V and
SM, Donato J Jr, Carpinelli AR, Abdulkader F. Zinc supplementation
Yadav UCS. New Delhi: Springer India, 2015, p. 17–29.
improves glucose homeostasis in high fat-fed mice by enhancing pan-
8. Boden MJ, Brandon AE, Tid-Ang JD, Preston E, Wilks D, Stuart E,
creatic ␤-cell function. Nutrients 9: 1150, 2017. doi:10.3390/nu9101150.
Cleasby ME, Turner N, Cooney GJ, Kraegen EW. Overexpression of
26. Copps KD, White MF. Regulation of insulin sensitivity by serine/
manganese superoxide dismutase ameliorates high-fat diet-induced insu-
lin resistance in rat skeletal muscle. Am J Physiol Endocrinol Metab 303: threonine phosphorylation of insulin receptor substrate proteins IRS1 and
E798 –E805, 2012. doi:10.1152/ajpendo.00577.2011. IRS2. Diabetologia 55: 2565–2582, 2012. doi:10.1007/s00125-012-
9. Bonnard C, Durand A, Peyrol S, Chanseaume E, Chauvin MA, 2644-8.
Morio B, Vidal H, Rieusset J. Mitochondrial dysfunction results from 27. Corbett JA, Sweetland MA, Lancaster JR Jr, McDaniel ML. A
oxidative stress in the skeletal muscle of diet-induced insulin-resistant 1-hour pulse with IL-1 beta induces formation of nitric oxide and inhibits
mice. J Clin Invest 118: 789 –800, 2008. doi:10.1172/JCI32601. insulin secretion by rat islets of Langerhans: evidence for a tyrosine
10. Boshtam M, Rafiei M, Golshadi ID, Ani M, Shirani Z, Rostamshirazi kinase signaling mechanism. FASEB J 7: 369 –374, 1993. doi:10.1096/
M. Long term effects of oral vitamin E supplement in type II diabetic fasebj.7.2.8440413.
patients. Int J Vitam Nutr Res 75: 341–346, 2005. doi:10.1024/0300- 28. Corless M, Kiely A, McClenaghan NH, Flatt PR, Newsholme P.
9831.75.5.341. Glutamine regulates expression of key transcription factor, signal trans-
11. Boyle KE, Newsom SA, Janssen RC, Lappas M, Friedman JE. duction, metabolic gene, and protein expression in a clonal pancreatic
Skeletal muscle MnSOD, mitochondrial complex II, and SIRT3 enzyme ␤-cell line. J Endocrinol 190: 719 –727, 2006. doi:10.1677/joe.1.06892.
activities are decreased in maternal obesity during human pregnancy and 29. Crow JP, Beckman JS. The role of peroxynitrite in nitric oxide-
gestational diabetes mellitus. J Clin Endocrinol Metab 98: E1601– mediated toxicity. Curr Top Microbiol Immunol 196: 57–73, 1995.
E1609, 2013. doi:10.1210/jc.2013-1943. doi:10.1007/978-3-642-79130-7_7.
12. Broniowska KA, Oleson BJ, McGraw J, Naatz A, Mathews CE, 30. Cruzat VF, Keane KN, Scheinpflug AL, Cordeiro R, Soares MJ,
Corbett JA. How the location of superoxide generation influences the Newsholme P. Alanyl-glutamine improves pancreatic ␤-cell function
␤-cell response to nitric oxide. J Biol Chem 290: 7952–7960, 2015. following ex vivo inflammatory challenge. J Endocrinol 224: 261–271,
doi:10.1074/jbc.M114.627869. 2015. doi:10.1530/JOE-14-0677.
13. Brownlee M, Cerami A. The biochemistry of the complications of 31. Cruzat VF, Pantaleão LC, Donato J Jr, de Bittencourt PI Jr,
diabetes mellitus. Annu Rev Biochem 50: 385–432, 1981. doi:10.1146/ Tirapegui J. Oral supplementations with free and dipeptide forms of
annurev.bi.50.070181.002125. L-glutamine in endotoxemic mice: effects on muscle glutamine-glutathi-

AJP-Cell Physiol • doi:10.1152/ajpcell.00141.2019 • www.ajpcell.org


Downloaded from journals.physiology.org/journal/ajpcell (189.239.040.188) on May 14, 2022.
C430 OXIDATIVE STRESS, CELL METABOLISM AND FUNCTION

one axis and heat shock proteins. J Nutr Biochem 25: 345–352, 2014. 51. Gao H, Geng T, Huang T, Zhao Q. Fish oil supplementation and insulin
doi:10.1016/j.jnutbio.2013.11.009. sensitivity: a systematic review and meta-analysis. Lipids Health Dis 16:
32. Cruzat VF, Tirapegui J. Effects of oral supplementation with glutamine 131, 2017. doi:10.1186/s12944-017-0528-0.
and alanyl-glutamine on glutamine, glutamate, and glutathione status in 52. Gehrmann W, Elsner M, Lenzen S. Role of metabolically generated
trained rats and subjected to long-duration exercise. Nutrition 25: 428 – reactive oxygen species for lipotoxicity in pancreatic ␤-cells. Diabetes
435, 2009. doi:10.1016/j.nut.2008.09.014. Obes Metab 12, Suppl 2: 149 –158, 2010. doi:10.1111/j.1463-1326.2010.
33. Dakhale GN, Chaudhari HV, Shrivastava M. Supplementation of 01265.x.
vitamin C reduces blood glucose and improves glycosylated hemoglobin 53. Gerber PA, Rutter GA. The role of oxidative stress and hypoxia in
in type 2 diabetes mellitus: a randomized, double-blind study. Adv pancreatic ␤-cell dysfunction in diabetes mellitus. Antioxid Redox Signal
Pharmacol Sci 2011: 1–5, 2011. doi:10.1155/2011/195271. 26: 501–518, 2017. doi:10.1089/ars.2016.6755.
34. David JA, Rifkin WJ, Rabbani PS, Ceradini DJ. The Nrf2/Keap1/ 54. Ghafourifar P, Cadenas E. Mitochondrial nitric oxide synthase. Trends
ARE pathway and oxidative stress as a therapeutic target in type II Pharmacol Sci 26: 190 –195, 2005. doi:10.1016/j.tips.2005.02.005.
diabetes mellitus. J Diabetes Res 2017: 1–15, 2017. doi:10.1155/2017/ 55. Giordano CR, Roberts R, Krentz KA, Bissig D, Talreja D, Kumar A,
4826724. Terlecky SR, Berkowitz BA. Catalase therapy corrects oxidative stress-
35. de Alvaro C, Teruel T, Hernandez R, Lorenzo M. Tumor necrosis induced pathophysiology in incipient diabetic retinopathy. Invest Oph-
factor alpha produces insulin resistance in skeletal muscle by activation thalmol Vis Sci 56: 3095–3102, 2015. doi:10.1167/iovs.14-16194.
of inhibitor kappaB kinase in a p38 MAPK-dependent manner. J Biol 56. Gloire G, Piette J. Redox regulation of nuclear post-translational mod-
Chem 279: 17070 –17078, 2004. doi:10.1074/jbc.M312021200. ifications during NF-kappaB activation. Antioxid Redox Signal 11: 2209 –
36. de Oliveira-Marques V, Cyrne L, Marinho HS, Antunes F. A quan- 2222, 2009. doi:10.1089/ars.2009.2463.
titative study of NF-␬B activation by H2O2: relevance in inflammation 57. Gold R, Kappos L, Arnold DL, Bar-Or A, Giovannoni G, Selmaj
and synergy with TNF-␣. J Immunol 178: 3893–3902, 2007. doi:10. K, Tornatore C, Sweetser MT, Yang M, Sheikh SI, Dawson KT;
4049/jimmunol.178.6.3893. DEFINE Study Investigators. Placebo-controlled phase 3 study of
37. Delaney CA, Tyrberg B, Bouwens L, Vaghef H, Hellman B, Eizirik oral BG-12 for relapsing multiple sclerosis. N Engl J Med 367: 1098 –
DL. Sensitivity of human pancreatic islets to peroxynitrite-induced cell 1107, 2012. doi:10.1056/NEJMoa1114287.
dysfunction and death. FEBS Lett 394: 300 –306, 1996. doi:10.1016/ 58. Gopaul NK, Manraj MD, Hébé A, Lee Kwai Yan S, Johnston A,
0014-5793(96)00977-5. Carrier MJ, Anggård EE. Oxidative stress could precede endothelial
38. Di Giacomo G, Rizza S, Montagna C, Filomeni G. Established prin- dysfunction and insulin resistance in Indian Mauritians with impaired
ciples and emerging concepts on the interplay between mitochondrial glucose metabolism. Diabetologia 44: 706 –712, 2001. doi:10.1007/
physiology and S-(de)nitrosylation: implications in cancer and neurode- s001250051679.
generation. Int J Cell Biol 2012: 1–20, 2012. doi:10.1155/2012/361872. 59. Góth L. A new type of inherited catalase deficiencies: its characteriza-
39. Di Naso FC, Simões Dias A, Porawski M, Marroni NA. Exogenous tion and comparison to the Japanese and Swiss type of acatalasemia.
superoxide dismutase: action on liver oxidative stress in animals with Blood Cells Mol Dis 27: 512–517, 2001. doi:10.1006/bcmd.2001.0415.
streptozotocin-induced diabetes. Exp Diabetes Res 2011: 1–6, 2011. 60. Góth L, Eaton JW. Hereditary catalase deficiencies and increased risk
doi:10.1155/2011/754132. of diabetes. Lancet 356: 1820 –1821, 2000. doi:10.1016/S0140-6736(00)
40. Dias IH, Griffiths HR. Oxidative stress in diabetes— circulating ad- 03238-4.
vanced glycation end products, lipid oxidation and vascular disease. Ann 61. Góth L, Lenkey A, Bigler WN. Blood catalase deficiency and diabetes
Clin Biochem 51: 125–127, 2014. doi:10.1177/0004563213508747. in Hungary. Diabetes Care 24: 1839 –1840, 2001. doi:10.2337/diacare.
41. Draznin B. Molecular mechanisms of insulin resistance: serine phos- 24.10.1839.
phorylation of insulin receptor substrate-1 and increased expression of 62. Graciano MF, Valle MM, Curi R, Carpinelli AR. Evidence for the
p85alpha: the two sides of a coin. Diabetes 55: 2392–2397, 2006. involvement of GPR40 and NADPH oxidase in palmitic acid-induced
doi:10.2337/db06-0391. superoxide production and insulin secretion. Islets 5: 139 –148, 2013.
42. Elsner M, Gehrmann W, Lenzen S. Peroxisome-generated hydrogen doi:10.4161/isl.25459.
peroxide as important mediator of lipotoxicity in insulin-producing cells. 63. Gray JP, Heart E. Usurping the mitochondrial supremacy: extramito-
Diabetes 60: 200 –208, 2011. doi:10.2337/db09-1401. chondrial sources of reactive oxygen intermediates and their role in
43. Evans JL, Goldfine ID, Maddux BA, Grodsky GM. Are oxidative ␤-cell metabolism and insulin secretion. Toxicol Mech Methods 20:
stress-activated signaling pathways mediators of insulin resistance and 167–174, 2010. doi:10.3109/15376511003695181.
␤-cell dysfunction? Diabetes 52: 1–8, 2003. doi:10.2337/diabetes.52.1.1. 64. Gual P, Le Marchand-Brustel Y, Tanti JF. Positive and negative
44. Fabre O, Breuker C, Amouzou C, Salehzada T, Kitzmann M, regulation of insulin signaling through IRS-1 phosphorylation. Biochimie
Mercier J, Bisbal C. Defects in TLR3 expression and RNase L activa- 87: 99 –109, 2005. doi:10.1016/j.biochi.2004.10.019.
tion lead to decreased MnSOD expression and insulin resistance in 65. Guo S, Dai C, Guo M, Taylor B, Harmon JS, Sander M, Robertson
muscle cells of obese people. Cell Death Dis 5: e1136, 2014. doi:10. RP, Powers AC, Stein R. Inactivation of specific ␤ cell transcription
1038/cddis.2014.104. factors in type 2 diabetes. J Clin Invest 123: 3305–3316, 2013. doi:10.
45. Finaud J, Lac G, Filaire E. Oxidative stress : relationship with exercise 1172/JCI65390.
and training. Sports Med 36: 327–358, 2006. doi:10.2165/00007256- 66. Gurgul-Convey E, Hanzelka K, Lenzen S. Is there a role for neuronal
200636040-00004. nitric oxide synthase (nNOS) in cytokine toxicity to pancreatic ␤-cells?
46. Finkel T. Reactive oxygen species and signal transduction. IUBMB Life Nitric Oxide 27: 235–241, 2012. doi:10.1016/j.niox.2012.08.075.
52: 3–6, 2001. doi:10.1080/15216540252774694. 67. Gurgul-Convey E, Mehmeti I, Lortz S, Lenzen S. Cytokine toxicity in
47. Fläring UB, Rooyackers OE, Wernerman J, Hammarqvist F. Glu- insulin-producing cells is mediated by nitro-oxidative stress-induced
tamine attenuates post-traumatic glutathione depletion in human muscle. hydroxyl radical formation in mitochondria. J Mol Med (Berl) 89:
Clin Sci (Lond) 104: 275–282, 2003. doi:10.1042/CS20020198. 785–798, 2011. doi:10.1007/s00109-011-0747-1.
48. Foster JR, Idle JR, Hardwick JP, Bars R, Scott P, Braganza JM. 68. Han W, Li H, Cai J, Gleaves LA, Polosukhin VV, Segal BH, Yull FE,
Induction of drug-metabolizing enzymes in human pancreatic cancer and Blackwell TS. NADPH oxidase limits lipopolysaccharide-induced lung
chronic pancreatitis. J Pathol 169: 457–463, 1993. doi:10.1002/path. inflammation and injury in mice through reduction-oxidation regulation
1711690412. of NF-␬B activity. J Immunol 190: 4786 –4794, 2013. doi:10.4049/
49. Fujishiro M, Gotoh Y, Katagiri H, Sakoda H, Ogihara T, Anai M, jimmunol.1201809.
Onishi Y, Ono H, Abe M, Shojima N, Fukushima Y, Kikuchi M, Oka 69. Hayes JD, McMahon M. The double-edged sword of Nrf2: subversion
Y, Asano T. Three mitogen-activated protein kinases inhibit insulin of redox homeostasis during the evolution of cancer. Mol Cell 21:
signaling by different mechanisms in 3T3-L1 adipocytes. Mol Endocri- 732–734, 2006. doi:10.1016/j.molcel.2006.03.004.
nol 17: 487–497, 2003. doi:10.1210/me.2002-0131. 70. He HJ, Wang GY, Gao Y, Ling WH, Yu ZW, Jin TR. Curcumin
50. Gao D, Nong S, Huang X, Lu Y, Zhao H, Lin Y, Man Y, Wang S, attenuates Nrf2 signaling defect, oxidative stress in muscle and glucose
Yang J, Li J. The effects of palmitate on hepatic insulin resistance are intolerance in high fat diet-fed mice. World J Diabetes 3: 94 –104, 2012.
mediated by NADPH Oxidase 3-derived reactive oxygen species through doi:10.4239/wjd.v3.i5.94.
JNK and p38MAPK pathways. J Biol Chem 285: 29965–29973, 2010. 71. Hebels DGAJ, Briedé JJ, Khampang R, Kleinjans JCS, de Kok
doi:10.1074/jbc.M110.128694. TMCM. Radical mechanisms in nitrosamine- and nitrosamide-induced

AJP-Cell Physiol • doi:10.1152/ajpcell.00141.2019 • www.ajpcell.org


Downloaded from journals.physiology.org/journal/ajpcell (189.239.040.188) on May 14, 2022.
OXIDATIVE STRESS, CELL METABOLISM AND FUNCTION C431
whole-genome gene expression modulations in Caco-2 cells. Toxicol Sci 90. Koshkin V, Wang X, Scherer PE, Chan CB, Wheeler MB. Mito-
116: 194 –205, 2010. doi:10.1093/toxsci/kfq121. chondrial functional state in clonal pancreatic ␤-cells exposed to free
72. Hernandez-Perez M, Chopra G, Fine J, Conteh AM, Anderson RM, fatty acids. J Biol Chem 278: 19709 –19715, 2003. doi:10.1074/jbc.
Linnemann AK, Benjamin C, Nelson JB, Benninger KS, Nadler JL, M209709200.
Maloney DJ, Tersey SA, Mirmira RG. Inhibition of 12/15-lipoxygen- 91. Koulajian K, Desai T, Liu GC, Ivovic A, Patterson JN, Tang C,
ase protects against ␤-cell oxidative stress and glycemic deterioration in El-Benna J, Joseph JW, Scholey JW, Giacca A. NADPH oxidase
mouse models of type 1 diabetes. Diabetes 66: 2875–2887, 2017. inhibition prevents ␤-cell dysfunction induced by prolonged elevation of
doi:10.2337/db17-0215. oleate in rodents. Diabetologia 56: 1078 –1087, 2013. doi:10.1007/
73. Homem de Bittencourt PI Jr, Curi R. Antiproliferative prostaglandins s00125-013-2858-4.
and the MRP/GS-X pump role in cancer immunosuppression and insight 92. Koulajian K, Ivovic A, Ye K, Desai T, Shah A, Fantus IG, Ran Q,
into new strategies in cancer gene therapy. Biochem Pharmacol 62: Giacca A. Overexpression of glutathione peroxidase 4 prevents ␤-cell
811–819, 2001. doi:10.1016/S0006-2952(01)00738-9. dysfunction induced by prolonged elevation of lipids in vivo. Am J
74. Houstis N, Rosen ED, Lander ES. Reactive oxygen species have a Physiol Endocrinol Metab 305: E254 –E262, 2013. doi:10.1152/ajpendo.
causal role in multiple forms of insulin resistance. Nature 440: 944 –948, 00481.2012.
2006. doi:10.1038/nature04634. 93. Kurrer MO, Pakala SV, Hanson HL, Katz JD. 〉-cell apoptosis in T
75. Hrycay EG, Bandiera SM. Chapter Two. Involvement of cytochrome cell-mediated autoimmune diabetes. Proc Natl Acad Sci USA 94: 213–
P450 in reactive oxygen species formation and cancer. In: Advances in 218, 1997. doi:10.1073/pnas.94.1.213.
Pharmacology, edited by Hardwick JP. Cambridge, MA: Academic, 94. Lajoix AD, Reggio H, Chardès T, Péraldi-Roux S, Tribillac F, Roye
2015, p. 35–84. M, Dietz S, Broca C, Manteghetti M, Ribes G, Wollheim CB, Gross
76. Huang JQ, Zhou JC, Wu YY, Ren FZ, Lei XG. Role of glutathione R. A neuronal isoform of nitric oxide synthase expressed in pancreatic
peroxidase 1 in glucose and lipid metabolism-related diseases. Free ␤-cells controls insulin secretion. Diabetes 50: 1311–1323, 2001. [Erra-
Radic Biol Med 127: 108 –115, 2018. doi:10.1016/j.freeradbiomed.2018. tum in Diabetes 50: 2177–2178, 2001.] doi:10.2337/diabetes.50.6.1311.
05.077. 95. Lee HY, Choi CS, Birkenfeld AL, Alves TC, Jornayvaz FR, Jurczak
77. Imoto H, Sasaki N, Iwase M, Nakamura U, Oku M, Sonoki K, MJ, Zhang D, Woo DK, Shadel GS, Ladiges W, Rabinovitch PS,
Uchizono Y, Iida M. Impaired insulin secretion by diphenyleneiodium Santos JH, Petersen KF, Samuel VT, Shulman GI. Targeted expres-
associated with perturbation of cytosolic Ca2⫹ dynamics in pancreatic sion of catalase to mitochondria prevents age-associated reductions in
␤-cells. Endocrinology 149: 5391–5400, 2008. doi:10.1210/en.2008- mitochondrial function and insulin resistance. Cell Metab 12: 668 –674,
0186. 2010. doi:10.1016/j.cmet.2010.11.004.
78. Ivarsson R, Quintens R, Dejonghe S, Tsukamoto K, in ’t Veld P, 96. Lees Murdock DJ, Barnett YA, Barnett CR. DNA damage and
Renström E, Schuit FC. Redox control of exocytosis: regulatory role of cytotoxicity in pancreatic ␤-cells expressing human CYP2E1. Biochem
NADPH, thioredoxin, and glutaredoxin. Diabetes 54: 2132–2142, 2005. Pharmacol 68: 523–530, 2004. doi:10.1016/j.bcp.2004.04.008.
doi:10.2337/diabetes.54.7.2132. 97. Leloup C, Tourrel-Cuzin C, Magnan C, Karaca M, Castel J, Car-
79. Jacobson J, Duchen MR. Interplay between mitochondria and cellular neiro L, Colombani AL, Ktorza A, Casteilla L, Pénicaud L. Mito-
calcium signalling. Mol Cell Biochem 256: 209 –218, 2004. doi:10.1023/ chondrial reactive oxygen species are obligatory signals for glucose-
B:MCBI.0000009869.29827.df. induced insulin secretion. Diabetes 58: 673–681, 2009. doi:10.2337/
80. Jiang T, Huang Z, Lin Y, Zhang Z, Fang D, Zhang DD. The protective db07-1056.
role of Nrf2 in streptozotocin-induced diabetic nephropathy. Diabetes 59: 98. Lenzen S, Drinkgern J, Tiedge M. Low antioxidant enzyme gene
850 –860, 2010. doi:10.2337/db09-1342. expression in pancreatic islets compared with various other mouse
81. Jiménez-Osorio AS, González-Reyes S, Pedraza-Chaverri J. Natural tissues. Free Radic Biol Med 20: 463–466, 1996. doi:10.1016/0891-
Nrf2 activators in diabetes. Clin Chim Acta 448: 182–192, 2015. doi:10. 5849(96)02051-5.
1016/j.cca.2015.07.009. 99. Lepretti M, Martucciello S, Burgos Aceves MA, Putti R, Lionetti L.
82. Jing E, Emanuelli B, Hirschey MD, Boucher J, Lee KY, Lombard D, Omega-3 fatty acids and insulin resistance: focus on the regulation of
Verdin EM, Kahn CR. Sirtuin-3 (Sirt3) regulates skeletal muscle mitochondria and endoplasmic reticulum stress. Nutrients 10: 350, 2018.
metabolism and insulin signaling via altered mitochondrial oxidation and doi:10.3390/nu10030350.
reactive oxygen species production. Proc Natl Acad Sci USA 108: 100. Lewandowski L, Kepinska M, Milnerowicz H. The copper-zinc super-
14608 –14613, 2011. doi:10.1073/pnas.1111308108. oxide dismutase activity in selected diseases. Eur J Clin Invest 49:
83. Kabe Y, Ando K, Hirao S, Yoshida M, Handa H. Redox regulation of e13036, 2018. doi:10.1111/eci.13036.
NF-kappaB activation: distinct redox regulation between the cytoplasm 101. Lin N, Chen H, Zhang H, Wan X, Su Q. Mitochondrial reactive oxygen
and the nucleus. Antioxid Redox Signal 7: 395–403, 2005. doi:10.1089/ species (ROS) inhibition ameliorates palmitate-induced INS-1 ␤-cell
ars.2005.7.395. death. Endocrine 42: 107–117, 2012. doi:10.1007/s12020-012-9633-z.
84. Kaneto H, Katakami N, Matsuhisa M, Matsuoka TA. Role of reactive 102. Loh K, Deng H, Fukushima A, Cai X, Boivin B, Galic S, Bruce C,
oxygen species in the progression of type 2 diabetes and atherosclerosis. Shields BJ, Skiba B, Ooms LM, Stepto N, Wu B, Mitchell CA, Tonks
Mediators Inflamm 2010: 1–11, 2010. doi:10.1155/2010/453892. NK, Watt MJ, Febbraio MA, Crack PJ, Andrikopoulos S, Tiganis T.
85. Kaneto H, Xu G, Fujii N, Kim S, Bonner-Weir S, Weir GC. Involve- Reactive oxygen species enhance insulin sensitivity. Cell Metab 10:
ment of c-Jun N-terminal kinase in oxidative stress-mediated suppression 260 –272, 2009. doi:10.1016/j.cmet.2009.08.009.
of insulin gene expression. J Biol Chem 277: 30010 –30018, 2002. 103. Lortz S, Gurgul-Convey E, Lenzen S, Tiedge M. Importance of
doi:10.1074/jbc.M202066200. mitochondrial superoxide dismutase expression in insulin-producing
86. Kawamori D, Kaneto H, Nakatani Y, Matsuoka TA, Matsuhisa M, cells for the toxicity of reactive oxygen species and proinflammatory
Hori M, Yamasaki Y. The forkhead transcription factor Foxo1 bridges cytokines. Diabetologia 48: 1541–1548, 2005. doi:10.1007/s00125-005-
the JNK pathway and the transcription factor PDX-1 through its intra- 1822-3.
cellular translocation. J Biol Chem 281: 1091–1098, 2006. doi:10.1074/ 104. Lucas D, Farez C, Bardou LG, Vaisse J, Attali JR, Valensi P.
jbc.M508510200. Cytochrome P450 2E1 activity in diabetic and obese patients as assessed
87. Kikuchi N, Ishii Y, Morishima Y, Yageta Y, Haraguchi N, Itoh K, by chlorzoxazone hydroxylation. Fundam Clin Pharmacol 12: 553–558,
Yamamoto M, Hizawa N. Nrf2 protects against pulmonary fibrosis by 1998. doi:10.1111/j.1472-8206.1998.tb00985.x.
regulating the lung oxidant level and Th1/Th2 balance. Respir Res 11: 31, 105. Madhumitha H, Mohan V, Deepa M, Babu S, Aravindhan V. In-
2010. doi:10.1186/1465-9921-11-31. creased Th1 and suppressed Th2 serum cytokine levels in subjects with
88. Kim JK. Hyperinsulinemic-euglycemic clamp to assess insulin sensitiv- diabetic coronary artery disease. Cardiovasc Diabetol 13: 1, 2014.
ity in vivo. Methods Mol Biol 560: 221–238, 2009. doi:10.1007/978-1- doi:10.1186/1475-2840-13-1.
59745-448-3_15. 106. Mancini A, Martorana GE, Magini M, Festa R, Raimondo S, Silves-
89. Kopprasch S, Srirangan D, Bergmann S, Graessler J, Schwarz PEH, trini A, Nicolotti N, Mordente A, Mele MC, Miggiano GAD, Meucci
Bornstein SR. Association between systemic oxidative stress and insulin E. Oxidative stress and metabolic syndrome: Effects of a natural antiox-
resistance/sensitivity indices - the PREDIAS study. Clin Endocrinol idants enriched diet on insulin resistance. Clin Nutr ESPEN 10: e52–e60,
(Oxf) 84: 48 –54, 2016. doi:10.1111/cen.12811. 2015. doi:10.1016/j.clnesp.2014.11.002.

AJP-Cell Physiol • doi:10.1152/ajpcell.00141.2019 • www.ajpcell.org


Downloaded from journals.physiology.org/journal/ajpcell (189.239.040.188) on May 14, 2022.
C432 OXIDATIVE STRESS, CELL METABOLISM AND FUNCTION

107. Manning PJ, Sutherland WH, Walker RJ, Williams SM, De Jong 124. Nishikawa T, Edelstein D, Du XL, Yamagishi S, Matsumura T,
SA, Ryalls AR, Berry EA. Effect of high-dose vitamin E on insulin Kaneda Y, Yorek MA, Beebe D, Oates PJ, Hammes HP, Giardino I,
resistance and associated parameters in overweight subjects. Diabetes Brownlee M. Normalizing mitochondrial superoxide production blocks
Care 27: 2166 –2171, 2004. doi:10.2337/diacare.27.9.2166. three pathways of hyperglycaemic damage. Nature 404: 787–790, 2000.
108. Mansuroğlu B, Derman S, Yaba A, Kızılbey K. Protective effect of doi:10.1038/35008121.
chemically modified SOD on lipid peroxidation and antioxidant status in 125. Nishimura W, Kondo T, Salameh T, El Khattabi I, Dodge R,
diabetic rats. Int J Biol Macromol 72: 79 –87, 2015. doi:10.1016/j. Bonner-Weir S, Sharma A. A switch from MafB to MafA expression
ijbiomac.2014.07.039. accompanies differentiation to pancreatic ␤-cells. Dev Biol 293: 526 –
109. Matsuzawa-Nagata N, Takamura T, Ando H, Nakamura S, Kurita S, 539, 2006. doi:10.1016/j.ydbio.2006.02.028.
Misu H, Ota T, Yokoyama M, Honda M, Miyamoto K, Kaneko S. 126. Nukatsuka M, Sakurai H, Yoshimura Y, Nishida M, Kawada J.
Increased oxidative stress precedes the onset of high-fat diet-induced Enhancement by streptozotocin of O2- radical generation by the xanthine
insulin resistance and obesity. Metabolism 57: 1071–1077, 2008. doi:10. oxidase system of pancreatic ␤-cells. FEBS Lett 239: 295–298, 1988.
1016/j.metabol.2008.03.010. doi:10.1016/0014-5793(88)80938-4.
110. Meigs JB, Larson MG, Fox CS, Keaney JF Jr, Vasan RS, Benjamin 127. Ogasawara A, Simizu S, Ito A, Kawai T, Saisho Y, Takei I, Umezawa
EJ. Association of oxidative stress, insulin resistance, and diabetes risk K. Inhibition of NO-induced ␤-cell death by novel NF-␬B inhibitor
phenotypes: the Framingham Offspring Study. Diabetes Care 30: 2529 – (–)-DHMEQ via activation of Nrf2-ARE pathway. Biochem Biophys Res
2535, 2007. doi:10.2337/dc07-0817. Commun 433: 181–187, 2013. doi:10.1016/j.bbrc.2013.02.062.
111. Menge BA, Schrader H, Ritter PR, Ellrichmann M, Uhl W, Schmidt 128. Ogawa-Wong AN, Berry MJ, Seale LA. Selenium and metabolic
WE, Meier JJ. Selective amino acid deficiency in patients with impaired disorders: an emphasis on type 2 diabetes risk. Nutrients 8: 80, 2016.
glucose tolerance and type 2 diabetes. Regul Pept 160: 75–80, 2010. doi:10.3390/nu8020080.
doi:10.1016/j.regpep.2009.08.001. 129. Oliveira HR, Verlengia R, Carvalho CRO, Britto LRG, Curi R,
112. Merry TL, Tran M, Stathopoulos M, Wiede F, Fam BC, Dodd GT, Carpinelli AR. Pancreatic ␤-cells express phagocyte-like NAD(P)H
Clarke I, Watt MJ, Andrikopoulos S, Tiganis T. High-fat-fed obese oxidase. Diabetes 52: 1457–1463, 2003. doi:10.2337/diabetes.52.6.1457.
glutathione peroxidase 1-deficient mice exhibit defective insulin secre- 130. Opara EC, Abdel-Rahman E, Soliman S, Kamel WA, Souka S, Lowe
tion but protection from hepatic steatosis and liver damage. Antioxid JE, Abdel-Aleem S. Depletion of total antioxidant capacity in type 2
Redox Signal 20: 2114 –2129, 2014. doi:10.1089/ars.2013.5428. diabetes. Metabolism 48: 1414 –1417, 1999. doi:10.1016/S0026-0495
113. Miki A, Ricordi C, Sakuma Y, Yamamoto T, Misawa R, Mita A, (99)90152-X.
Molano RD, Vaziri ND, Pileggi A, Ichii H. Divergent antioxidant 131. Padgett LE, Broniowska KA, Hansen PA, Corbett JA, Tse HM. The
capacity of human islet cell subsets: A potential cause of ␤-cell vulner- role of reactive oxygen species and proinflammatory cytokines in type 1
ability in diabetes and islet transplantation. PLoS One 13: e0196570, diabetes pathogenesis. Ann N Y Acad Sci 1281: 16 –35, 2013. doi:10.
2018. doi:10.1371/journal.pone.0196570. 1111/j.1749-6632.2012.06826.x.
114. Morgan D, Oliveira-Emilio HR, Keane D, Hirata AE, Santos da 132. Park K, Gross M, Lee DH, Holvoet P, Himes JH, Shikany JM,
Jacobs DR Jr. Oxidative stress and insulin resistance: the coronary
Rocha M, Bordin S, Curi R, Newsholme P, Carpinelli AR. Glucose,
artery risk development in young adults study. Diabetes Care 32:
palmitate and proinflammatory cytokines modulate production and ac-
1302–1307, 2009. doi:10.2337/dc09-0259.
tivity of a phagocyte-like NADPH oxidase in rat pancreatic islets and a
133. Park K, Steffes M, Lee DH, Himes JH, Jacobs DR Jr. Association of
clonal ␤-cell line. Diabetologia 50: 359 –369, 2007. doi:10.1007/s00125-
inflammation with worsening HOMA-insulin resistance. Diabetologia
006-0462-6.
52: 2337–2344, 2009. doi:10.1007/s00125-009-1486-5.
115. Morgan D, Rebelato E, Abdulkader F, Graciano MFR, Oliveira-
134. Phaniendra A, Jestadi DB, Periyasamy L. Free radicals: properties,
Emilio HR, Hirata AE, Rocha MS, Bordin S, Curi R, Carpinelli AR.
sources, targets, and their implication in various diseases. Indian J Clin
Association of NAD(P)H oxidase with glucose-induced insulin secretion
Biochem 30: 11–26, 2015. doi:10.1007/s12291-014-0446-0.
by pancreatic ␤-cells. Endocrinology 150: 2197–2201, 2009. doi:10.
135. Pi J, Bai Y, Zhang Q, Wong V, Floering LM, Daniel K, Reece JM,
1210/en.2008-1149. Deeney JT, Andersen ME, Corkey BE, Collins S. Reactive oxygen
116. Mugabo Y, Zhao S, Lamontagne J, Al-Mass A, Peyot M-L, Corkey species as a signal in glucose-stimulated insulin secretion. Diabetes 56:
BE, Joly E, Madiraju SRM, Prentki M. Metabolic fate of glucose and 1783–1791, 2007. doi:10.2337/db06-1601.
candidate signaling and excess-fuel detoxification pathways in pancreatic 136. Pi J, Leung L, Xue P, Wang W, Hou Y, Liu D, Yehuda-Shnaidman
␤-cells. J Biol Chem 292: 7407–7422, 2017. doi:10.1074/jbc.M116. E, Lee C, Lau J, Kurtz TW, Chan JY. Deficiency in the nuclear factor
763060. E2-related factor-2 transcription factor results in impaired adipogenesis
117. Munhoz AC, Riva P, Simões D, Curi R, Carpinelli AR. Control of and protects against diet-induced obesity. J Biol Chem 285: 9292–9300,
insulin secretion by production of reactive oxygen species: study per- 2010. doi:10.1074/jbc.M109.093955.
formed in pancreatic islets from fed and 48-hour fasted wistar rats. PLoS 137. Pullen TJ, Rutter GA. When less is more: the forbidden fruits of gene
One 11: e0158166, 2016. doi:10.1371/journal.pone.0158166. repression in the adult ␤-cell. Diabetes Obes Metab 15: 503–512, 2013.
118. Müssig K, Fiedler H, Staiger H, Weigert C, Lehmann R, Schleicher doi:10.1111/dom.12029.
ED, Häring HU. Insulin-induced stimulation of JNK and the PI 3-ki- 138. Quintana-Lopez L, Blandino-Rosano M, Perez-Arana G, Cebada-
nase/mTOR pathway leads to phosphorylation of serine 318 of IRS-1 in Aleu A, Lechuga-Sancho A, Aguilar-Diosdado M, Segundo C. Nitric
C2C12 myotubes. Biochem Biophys Res Commun 335: 819 –825, 2005. oxide is a mediator of antiproliferative effects induced by proinflamma-
doi:10.1016/j.bbrc.2005.07.154. tory cytokines on pancreatic ␤-cells. Mediators Inflamm 2013: 1–10,
119. Newsholme P, Cruzat V, Arfuso F, Keane K. Nutrient regulation of 2013. doi:10.1155/2013/905175.
insulin secretion and action. J Endocrinol 221: R105–R120, 2014. 139. Rabinovitch A, Suarez-Pinzon WL. Role of cytokines in the pathogen-
doi:10.1530/JOE-13-0616. esis of autoimmune diabetes mellitus. Rev Endocr Metab Disord 4:
120. Newsholme P, Cruzat VF, Keane KN, Carlessi R, de Bittencourt PIH 291–299, 2003. doi:10.1023/A:1025160614313.
Jr. Molecular mechanisms of ROS production and oxidative stress in 140. Radi R, Turrens JF, Chang LY, Bush KM, Crapo JD, Freeman BA.
diabetes. Biochem J 473: 4527–4550, 2016. doi:10.1042/BCJ20160503C. Detection of catalase in rat heart mitochondria. J Biol Chem 266:
121. Newsholme P, Morgan D, Rebelato E, Oliveira-Emilio HC, Procopio 22028 –22034, 1991.
J, Curi R, Carpinelli A. Insights into the critical role of NADPH 141. Ramprasath T, Kumar PH, Puhari SS, Murugan PS, Vasudevan V,
oxidase(s) in the normal and dysregulated pancreatic ␤-cell. Diabetolo- Selvam GS. L-Arginine ameliorates cardiac left ventricular oxidative
gia 52: 2489 –2498, 2009. doi:10.1007/s00125-009-1536-z. stress by upregulating eNOS and Nrf2 target genes in alloxan-induced
122. Newsholme P, Rebelato E, Abdulkader F, Krause M, Carpinelli A, hyperglycemic rats. Biochem Biophys Res Commun 428: 389 –394, 2012.
Curi R. Reactive oxygen and nitrogen species generation, antioxidant doi:10.1016/j.bbrc.2012.10.064.
defenses, and ␤-cell function: a critical role for amino acids. J Endocrinol 142. Rayman MP. Selenium and human health. Lancet 379: 1256 –1268,
214: 11–20, 2012. doi:10.1530/JOE-12-0072. 2012. doi:10.1016/S0140-6736(11)61452-9.
123. Nishikawa T, Edelstein D, Brownlee M. The missing link: a single 143. Rayman MP, Stranges S. Epidemiology of selenium and type 2 diabe-
unifying mechanism for diabetic complications. Kidney Int Suppl 58: tes: can we make sense of it? Free Radic Biol Med 65: 1557–1564, 2013.
S26 –S30, 2000. doi:10.1046/j.1523-1755.2000.07705.x. doi:10.1016/j.freeradbiomed.2013.04.003.

AJP-Cell Physiol • doi:10.1152/ajpcell.00141.2019 • www.ajpcell.org


Downloaded from journals.physiology.org/journal/ajpcell (189.239.040.188) on May 14, 2022.
OXIDATIVE STRESS, CELL METABOLISM AND FUNCTION C433
144. Rizzo MA, Piston DW. Regulation of ␤-cell glucokinase by S-nitrosy- 163. Trachootham D, Lu W, Ogasawara MA, Rivera-Del Valle N, Huang
lation and association with nitric oxide synthase. J Cell Biol 161: P. Redox regulation of cell survival. Antioxid Redox Signal 10: 1343–
243–248, 2003. doi:10.1083/jcb.200301063. 1374, 2008. doi:10.1089/ars.2007.1957.
145. Rockwell CE, Zhang M, Fields PE, Klaassen CD. Th2 skewing by 164. Udupa AS, Nahar PS, Shah SH, Kshirsagar MJ, Ghongane BB.
activation of Nrf2 in CD4(⫹) T cells. J Immunol 188: 1630 –1637, 2012. Study of comparative effects of antioxidants on insulin sensitivity in type
doi:10.4049/jimmunol.1101712. 2 diabetes mellitus. J Clin Diagn Res 6: 1469 –1473, 2012. doi:10.7860/
146. Rutten EP, Engelen MP, Schols AM, Deutz NE. Skeletal muscle JCDR/2012/4464.2535.
glutamate metabolism in health and disease: state of the art. Curr 165. Urakawa H, Katsuki A, Sumida Y, Gabazza EC, Murashima S,
Opin Clin Nutr Metab Care 8: 41–51, 2005. doi:10.1097/00075197- Morioka K, Maruyama N, Kitagawa N, Tanaka T, Hori Y, Nakatani
200501000-00007. K, Yano Y, Adachi Y. Oxidative stress is associated with adiposity and
147. Sampson SR, Bucris E, Horovitz-Fried M, Parnas A, Kahana S, insulin resistance in men. J Clin Endocrinol Metab 88: 4673–4676, 2003.
Abitbol G, Chetboun M, Rosenzweig T, Brodie C, Frankel S. Insulin doi:10.1210/jc.2003-030202.
increases H2O2-induced pancreatic ␤-cell death. Apoptosis 15: 1165– 166. Veal EA, Day AM, Morgan BA. Hydrogen peroxide sensing and
1176, 2010. doi:10.1007/s10495-010-0517-5. signaling. Mol Cell 26: 1–14, 2007. doi:10.1016/j.molcel.2007.03.016.
148. Sato Y, Fujimoto S, Mukai E, Sato H, Tahara Y, Ogura K, Yamano 167. Venoji R, Amirtharaj GJ, Kini A, Vanaparthi S, Venkatraman A,
G, Ogura M, Nagashima K, Inagaki N. Palmitate induces reactive Ramachandran A. Enteral glutamine differentially regulates Nrf 2 along
oxygen species production and ␤-cell dysfunction by activating nicotin- the villus-crypt axis of the intestine to enhance glutathione levels. J
amide adenine dinucleotide phosphate oxidase through Src signaling. J Gastroenterol Hepatol 30: 1740 –1747, 2015. doi:10.1111/jgh.13019.
Diabetes Investig 5: 19 –26, 2014. doi:10.1111/jdi.12124. 168. Vincent HK, Bourguignon CM, Weltman AL, Vincent KR, Barrett
149. Schoonbroodt S, Piette J. Oxidative stress interference with the nuclear E, Innes KE, Taylor AG. Effects of antioxidant supplementation on
factor-kappa B activation pathways. Biochem Pharmacol 60: 1075–1083, insulin sensitivity, endothelial adhesion molecules, and oxidative stress
2000. doi:10.1016/S0006-2952(00)00371-3. in normal-weight and overweight young adults. Metabolism 58: 254 –
150. Schuit F, De Vos A, Farfari S, Moens K, Pipeleers D, Brun T, Prentki 262, 2009. doi:10.1016/j.metabol.2008.09.022.
M. Metabolic fate of glucose in purified islet cells. Glucose-regulated 169. Wang J, Wang H. Oxidative stress in pancreatic ␤-cell regeneration.
anaplerosis in ␤ cells. J Biol Chem 272: 18572–18579, 1997. doi:10. Oxid Med Cell Longev 2017: 1–9, 2017. doi:10.1155/2017/1930261.
1074/jbc.272.30.18572. 170. Wang X, Vatamaniuk MZ, Roneker CA, Pepper MP, Hu LG,
151. Simon SF, Taylor CG. Dietary zinc supplementation attenuates hyper- Simmons RA, Lei XG. Knockouts of SOD1 and GPX1 exert different
glycemia in db/db mice. Exp Biol Med (Maywood) 226: 43–51, 2001. impacts on murine islet function and pancreatic integrity. Antioxid Redox
doi:10.1177/153537020122600107. Signal 14: 391–401, 2011. doi:10.1089/ars.2010.3302.
152. Sireesh D, Dhamodharan U, Ezhilarasi K, Vijay V, Ramkumar KM. 171. Wang Z, Hall SD, Maya JF, Li L, Asghar A, Gorski JC. Diabetes
Association of NF-E2 Related Factor 2 (Nrf2) and inflammatory cyto- mellitus increases the in vivo activity of cytochrome P450 2E1 in
kines in recent onset Type 2 Diabetes Mellitus. Sci Rep 8: 5126, 2018. humans. Br J Clin Pharmacol 55: 77–85, 2003. doi:10.1046/j.1365-2125.
doi:10.1038/s41598-018-22913-6. 2003.01731.x.
153. Smukler SR, Tang L, Wheeler MB, Salapatek AM. Exogenous nitric 172. Wijekoon EP, Skinner C, Brosnan ME, Brosnan JT. Amino acid
oxide and endogenous glucose-stimulated ␤-cell nitric oxide augment metabolism in the Zucker diabetic fatty rat: effects of insulin resistance
insulin release. Diabetes 51: 3450 –3460, 2002. doi:10.2337/diabetes.51. and of type 2 diabetes. Can J Physiol Pharmacol 82: 506 –514, 2004.
12.3450. doi:10.1139/y04-067.
154. Song M-Y, Kim E-K, Moon W-S, Park J-W, Kim H-J, So H-S, Park 173. Wiseman DA, Kalwat MA, Thurmond DC. Stimulus-induced S-ni-
R, Kwon K-B, Park B-H. Sulforaphane protects against cytokine- and trosylation of Syntaxin 4 impacts insulin granule exocytosis. J Biol Chem
streptozotocin-induced ␤-cell damage by suppressing the NF-kappaB 286: 16344 –16354, 2011. doi:10.1074/jbc.M110.214031.
pathway. Toxicol Appl Pharmacol 235: 57–67, 2009. doi:10.1016/j.taap. 174. Yagishita Y, Fukutomi T, Sugawara A, Kawamura H, Takahashi T,
2008.11.007. Pi J, Uruno A, Yamamoto M. Nrf2 protects pancreatic ␤-cells from
155. Stančić A, Otašević V, Janković A, Vučetić M, Ivanović-Burmazović oxidative and nitrosative stress in diabetic model mice. Diabetes 63:
I, Filipović MR, Korać A, Markelić M, Veličković K, Golić I, 605–618, 2014. doi:10.2337/db13-0909.
Buzadžić B, Korać B. Molecular basis of hippocampal energy metab- 175. Yin Q, Ma Y, Hong Y, Hou X, Chen J, Shen C, Sun M, Shang Y,
olism in diabetic rats: the effects of SOD mimic. Brain Res Bull 99: Dong S, Zeng Z, Pei JJ, Liu X. Lycopene attenuates insulin signaling
27–33, 2013. doi:10.1016/j.brainresbull.2013.09.009. deficits, oxidative stress, neuroinflammation, and cognitive impairment
156. Sun W, Yang J, Wang W, Hou J, Cheng Y, Fu Y, Xu Z, Cai L. The in fructose-drinking insulin resistant rats. Neuropharmacology 86: 389 –
beneficial effects of Zn on Akt-mediated insulin and cell survival sig- 396, 2014. doi:10.1016/j.neuropharm.2014.07.020.
naling pathways in diabetes. J Trace Elem Med Biol 46: 117–127, 2018. 176. Zeng Z, He W, Jia Z, Hao S. Lycopene improves insulin sensitivity
doi:10.1016/j.jtemb.2017.12.005. through inhibition of STAT3/Srebp-1c-mediated lipid accumulation and
157. Swaminathan S, Fonseca VA, Alam MG, Shah SV. The role of iron in inflammation in mice fed a high-fat diet. Exp Clin Endocrinol Diabetes
diabetes and its complications. Diabetes Care 30: 1926 –1933, 2007. 125: 610 –617, 2017. doi:10.1055/s-0043-101919.
doi:10.2337/dc06-2625. 177. Zhang L, Keung W, Samokhvalov V, Wang W, Lopaschuk GD. Role
158. Takahara S. Progressive oral gangrene probably due to lack of catalase of fatty acid uptake and fatty acid beta-oxidation in mediating insulin
in the blood (acatalasaemia); report of nine cases. Lancet 260: 1101– resistance in heart and skeletal muscle. Biochim Biophys Acta 1801:
1104, 1952. doi:10.1016/S0140-6736(52)90939-2. 1–22, 2010. doi:10.1016/j.bbalip.2009.09.014.
159. Takemoto K, Tanaka M, Iwata H, Nishihara R, Ishihara K, Wang 178. Zhang YK, Wu KC, Liu J, Klaassen CD. Nrf2 deficiency improves
DH, Ogino K, Taniuchi K, Masuoka N. Low catalase activity in blood glucose tolerance in mice fed a high-fat diet. Toxicol Appl Pharmacol
is associated with the diabetes caused by alloxan. Clin Chim Acta 407: 264: 305–314, 2012. doi:10.1016/j.taap.2012.09.014.
43–46, 2009. doi:10.1016/j.cca.2009.06.028. 179. Zhang Z, Li J, Yang L, Chen R, Yang R, Zhang H, Cai D, Chen H.
160. Tan Y, Ichikawa T, Li J, Si Q, Yang H, Chen X, Goldblatt CS, Meyer The cytotoxic role of intermittent high glucose on apoptosis and cell
CJ, Li X, Cai L, Cui T. Diabetic downregulation of Nrf2 activity via viability in pancreatic ␤-cells. J Diabetes Res 2014: 1–9, 2014. doi:10.
ERK contributes to oxidative stress-induced insulin resistance in cardiac 1155/2014/712781.
cells in vitro and in vivo. Diabetes 60: 625–633, 2011. doi:10.2337/db10- 180. Zhong Q, Mishra M, Kowluru RA. Transcription factor Nrf2-mediated
1164. antioxidant defense system in the development of diabetic retinopathy.
161. Tiedge M, Lortz S, Drinkgern J, Lenzen S. Relation between Invest Ophthalmol Vis Sci 54: 3941–3948, 2013. doi:10.1167/iovs.13-
antioxidant enzyme gene expression and antioxidative defense status 11598.
of insulin-producing cells. Diabetes 46: 1733–1742, 1997. doi:10. 181. Zhu X, Wu C, Qiu S, Yuan X, Li L. Effects of resveratrol on glucose
2337/diab.46.11.1733. control and insulin sensitivity in subjects with type 2 diabetes: systematic
162. Toledo FGS. Mitochondrial involvement in skeletal muscle insulin review and meta-analysis. Nutr Metab (Lond) 14: 60, 2017. doi:10.1186/
resistance. Diabetes 63: 59 –61, 2014. doi:10.2337/db13-1427. s12986-017-0217-z.

AJP-Cell Physiol • doi:10.1152/ajpcell.00141.2019 • www.ajpcell.org


Downloaded from journals.physiology.org/journal/ajpcell (189.239.040.188) on May 14, 2022.

You might also like