You are on page 1of 26

Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

Contents lists available at ScienceDirect

Journal of Pharmaceutical and Biomedical Analysis


journal homepage: www.elsevier.com/locate/jpba

Review

Biomarkers of Oxidative Stress and Antioxidant Defense


a
Sema Demirci-Çekiç , Gülay Özkan , Aslı Neslihan Avan , Seda Uzunboy , b a a
]]
]]]]]]
]]

⁎ ⁎⁎
Esra Çapanoğlu b, , Reşat Apak a,c,
a
Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpasa, Avcilar, 34320 Istanbul, Turkey
b
Department of Food Engineering, Faculty of Chemical and Metallurgical Engineering, Istanbul Technical Uviversity, Istanbul, Turkey
c
Turkish Academy of Sciences (TUBA), Vedat Dalokay St. No. 112, Cankaya, 06670 Ankara, Turkey

a r t i cl e i nfo a bstr ac t

Article history: A number of reactive oxygen and nitrogen species are produced during normal metabolism in human body.
Received 29 July 2021 These species can be both radical and non-radical and have varying degrees of reactivity. Although they
Received in revised form 27 October 2021 have some important functions in the human body, such as contributing to signal transmission and the
Accepted 11 November 2021
immune system, their presence must be balanced by the antioxidant defense system. The human body has
Available online 28 November 2021
an excellent intrinsic enzymatic antioxidant system in addition to different non-enzymatic antioxidants
having small molecular masses. An extrinsic source of antioxidants are foodstuffs such as fruits, vegetables,
Keywords:
Biomarkers herbs and spices, mostly rich in polyphenols. When the delicate biochemical balance between oxidants and
Oxidative stress antioxidants is disturbed in favor of oxidants, “oxidative stress” conditions emerge, under which reactive
Lipids species can cause oxidative damage to biomacromolecules such as proteins, carbohydrates, lipids and DNA.
Proteins This oxidative damage is often associated with cancer, aging, and neurodegenerative disorders. Because
DNA reactive species are extremely short-lived, it is almost impossible to measure their concentrations directly.
Antioxidant protection Although there are certain methods such as ESR / EPR that serve this purpose, they have some dis­
advantages and are quite costly systems. Therefore, products generated from oxidative damage of proteins,
lipids and DNA are often used to quantify the extent of oxidative damage rather than direct measurement of
reactive species. These oxidative damage products are usually known as biomarkers. Determination of the
concentrations of these biomarkers and changes in the concentration of protective antioxidants can provide
useful information for avoiding certain diseases and keep healthy conditions.
© 2021 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . 2


1.1. Reactive Oxygen Species (ROSs) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . 2
1.2. Antioxidants (AOXs) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . 3
2. Metabolic Oxidation Products . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . 4
2.1. Lipid Peroxidation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . 4
2.1.1. Lipid Peroxidation Products. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . 6
2.2. Protein Damage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . 9
2.2.1. Oxidation of the Protein Backbone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . 9
2.2.2. Oxidation of Amino Acids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . 9
2.2.3. Protein Carbonyl Derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... 10
2.3. DNA Damage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . 10
3. Antioxidants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... 10
3.1. Enzymatic Antioxidants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . 10
3.2. Non-Enzymatic Antioxidants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... 12
4. Analytical Strategies for the Measurement of Oxidative Stress and Related Antioxidant Defense . . . . . . ..... . ..... . ..... . ..... . ..... . ..... 15


Correspondence to: Istanbul Technical Uviversity, Department of Food Engineering, Faculty of Chemical and Metallurgical Engineering, Istanbul, Turkey.
⁎⁎
Correspondence to: Istanbul University-Cerrahpaşa, Department of Chemistry, Faculty of Engineering, Avcilar, 34320 Istanbul, Turkey.
E-mail addresses: capanogl@itu.edu.tr (E. Çapanoğlu), rapak@istanbul.edu.tr (R. Apak).

https://doi.org/10.1016/j.jpba.2021.114477
0731-7085/© 2021 Elsevier B.V. All rights reserved.
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

4.1. Determination of Lipid Oxidation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15


4.1.1. Determination of Lipid Hydroperoxides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
4.1.2. Determination of Thiobarbituric Acid Reactive
Substances (TBARS) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... . 16
4.2. Determination of Oxidative Protein Damage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... . 16
4.3. Determination of Oxidative DNA Damage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... . 17
5. Balance Between Oxidants and Antioxidants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... 18
6. Biomarkers of Oxidative Stress and Antioxidant
Defense in Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... 18
6.1. Breast cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... . 18
6.2. Prostate cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... . 19
6.3. Colorectal cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... . 19
6.4. Cervical cancer. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... . 19
6.5. Leukemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... . 19
7. Biomarkers of Oxidative Stress and Antioxidant Defense in Neurodegenerative Diseases . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... 19
7.1. Amyotrophic lateral sclerosis (ALS) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... . 19
7.2. Parkinson’s disease (PD) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... 20
7.3. Alzheimer’s disease (AD). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... 20
7.4. Autism spectrum disorder. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... 20
8. Biomarkers of Oxidative Stress and Antioxidant
Defense in Diabetes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... 20
8.1. Type 1 diabetes mellitus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... 20
8.2. Type 2 diabetes mellitus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... 21
8.3. Gestational diabetes mellitus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... 21
9. Conclusions and Perspectives. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... 21
Funding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... 21
Declaration of Competing Interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... 21
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..... . ..... . ..... . ..... . ..... . ..... . ..... 21

1. Introduction before redox biology [5]. In later years, addition of H2O2 to ery­
throcytes was expressed as “cells were subjected to oxidative stress”
1.1. Reactive Oxygen Species (ROSs) [6]. In 1985, the term began to be used to mean oxidative damage to
cells and organs as we understand it today [7]. In the chapter titled
A number of reactive oxygen−containing substances, including “Oxidative Stress: Introduction Descriptions” written by Sies (in the
free radicals such as hydroxyl (•OH) and superoxide (O2•-), and non- book named ‘Oxidative Stress’) this concept was formulated and
radical species such as hydrogen peroxide (H2O2), are commonly introduced as an imbalance between AOXs and oxidants in favor of
referred to as reactive oxygen species (ROS). There are also certain oxidants [8]. Later Sies improved his definition by adding an ex­
reactive species that carry different elements other than oxygen, pression which stated that oxidative stress causes a disruption on
such as nitrogen, sulfur and chlorine, so maybe it would be more redox signaling [9]. Considering the new definition of oxidative
appropriate to use the term “reactive species” (RS) instead of ROS. stress, this concept should be interpreted beyond the oxidative da­
Generation of RS is an unavoidable part of aerobic life due to the mage in biomolecules caused by free radicals and should be accepted
stepwise uptake of electrons by molecular oxygen from various as the disruption of the balanced cellular redox state [10]. However,
substrates, and RS can normally be balanced by antioxidants (AOX) the oxidative damage generated by RS in the macromolecules such
in a healthy organism [1,2]. Human beings have an immune system as DNA, lipid and protein is still an important topic. Damage gen­
that protects them from disease and keeps them alive, and RS have erated on these biological elements may result in cell death and
an active role to kill infecting pathogens. But after elimination of cause serious health problems. Different studies in the literature
threat, prolonging the activation of immune system also has harmful point out to a relationship between oxidative stress and some dis­
effects [3]. There are also various studies claiming that RS are ef­ eases such as cancer, atherosclerosis, Alzheimer’s disease (AD) and
fective in redox signaling in the body. It is also known that RS con­ Parkinson’s disease (PD). For example, it is known that cancer cells
tribute to adjustment for the situations caused by ischaemia and have higher amounts of RS compared to healthy cells. There is also
heavy exercise. To maintain a healthy life, the task of the human AOX some evidence indicating that protein carbonyls, generated at the
defense system is to eliminate excessive amounts of RS without end of oxidative protein damage, are present in Parkinson patients’
hindering them from maintaining their beneficial role [1]. The cel­ brain. There is also a link between oxidative stress and the biology of
lular AOX system tightly controls the amount of RS to maintain the Down syndrome. Chromosome 21, which is associated with Down
natural balance that exists in the cells, known as ‘redox home­ syndrome, is thought to have certain genes that contribute to neu­
ostasis’. The cellular AOX system consists of a series of components rodegeneration caused by oxidative stress. Other oxidative stress
including enzymatic and non-enzymatic species, mainly superoxide related health problems worth to mention are diabetes, chronic in­
dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx) as flammation and urolitiasis [11]. In addition, disruption of metal
enzymatic species and glutathione which is a non-enzymatic AOX. homeostasis can lead to various diseases. For example, Wilson’s
The function of SOD is catalyzing the dismutation reaction of O2•- disease and thalassemia are related to metal overload. The most
having end-products of O2 and H2O2. CAT and GPx decompose H2O2 likely reason for this is that an increase in metal ions in the body can
into H2O and O2. On the other hand, overproduction of RS cannot be trigger the formation of RS [12]. Thalassemia is a disorder associated
counterbalanced by AOX, leading to impairment of the balance in with defects in the synthesis of α- or β-globin subunits of he­
favor of oxidants and oxidative stress. High levels of oxidative stress moglobin A, i.e. these sub chains (α or β) can be reduced or absent.
may harm cellular macromolecules and increase cell death [4]. In­ Depending on the severity of the patient’s condition, a regular blood
terestingly, the term oxidative stress appeared in rubber chemistry transfusion may be needed. With or without a blood transfusion,

2
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

iron overload can be seen in thalassemia patients, and oral iron electron configuration of molecular oxygen (O2) shows that it is a
chelators may be used in disease management [13]. Although during diradical with 2 electrons singly occupying the two antibonding
the normal metabolism iron is bound to certain proteins, after orbitals at parallel spins. Thus, the relative chemical inertness of
several transfusions, iron binding proteins are saturated and free oxygen comes from this spin restriction, as a pair of electrons from a
iron (or non-transferrin bound iron) may accumulate in tissues or donor with antiparallel spins cannot be accommodated in the anti­
blood [14]. On the other hand, in thalassemias that do not require bonding orbitals of O2 each occupied by a single electron. In other
regular blood transfusion, the production of erythrocytes may cause words, oxygen takes up electrons one-by-one in stepwise manner,
an increase in gastrointestinal absorption and deposition of iron in initially leading to superoxide anion (as the rate-limiting step) fol­
the liver [15]. lowed by hydrogen peroxide [20]. If there are transition metal ions in
According to current knowledge it is a well-known fact that RS are the medium (especially at their lower oxidation states) where H2O2
routinely generated from different sources in most of the cells. Among is present, highly reactive radicals such as •OH are formed [21].
the endogenous sources of RS; mitochondria, oxidase enzymes (nico­ Additionally, H2O2 may directly attack heme proteins, inactivate
tinamide adenine dinucleotide phosphate (NADPH) oxidase, xanthine enzymes; oxidize -SH groups and biomacromolecules (e.g. DNA,
oxidase (XO), lipoxygenases, cyclooxygenases, polyamine and amino proteins and lipids) [22]. In the human body, H2O2 is consumed by
acid oxidases), other enzymes (cytochrome P450 enzymes, nitric oxide CAT. CAT converts H2O2 to water while it does not show this effect
synthases, flavin-dependent demethylase), heme groups, metal storage for other peroxides [23]. The hydroxyl radical, which can be pro­
proteins, free metal ions (iron and copper) can be mentioned. duced by the reaction between H2O2 and transition metal ions, is
Endogenously produced RS are consumed by enzymatic AOXs such as generally considered to be a highly reactive species that makes the
SOD, CAT, GPx and certain reductases. There are also different non- most important contribution to oxidative stress. For example, the
enzymatic, small molecular mass AOXs in the body such as ascorbate, Fenton reaction between Fe(II) and H2O2 which can be formulated as
pyruvate [16]. During aerobic respiration, ATP is produced to provide “Fe2+ + H2O2 → Fe3+ + •OH + OH-” is one of sources of hydroxyl ra­
the necessary energy for the body as a result of the reaction between dicals. Therefore, the type and amount of free and bound iron in the
oxygen and glucose in the mitochondria. In this process, O2 undergoes body is of particular importance. Iron is replenished as ferric ions in
step-by-step reduction and the generated intermediates are: hydro­ a Fenton reaction. O2•- is another RS in the body having the ability to
peroxyl radical (HOO•), superoxide anion radical (O2•-), hydrogen per­ release iron from iron-containing proteins such as ferritin and Fe-S
oxide (H2O2), hydroxyl radical (•OH), hydroxide anion (OH-), and the clusters. In normal metabolism, the level of iron is strictly regulated
final product is H2O: by metal-binding proteins. Only a small percentage of iron (i.e. less

According to this reduction scheme, oxygen is reduced by oxidase than 5% of total cell Fe) is present in the redox-active (labile) iron
enzymes to form superoxide anion radical (O2•-) which is formed in pool, essentially representing Fe-bound to low-affinity intracellular
mitochondrial electron transport chain during oxidative phosphoryla­ low-molecular-mass ligands [24]. However, the amount of catalytic
tion. While O2•- is a weak oxidant for ascorbic acid and thiols (such as iron increases under oxidative stress conditions [25], and this iron
reduced glutathione, GSH) in aqueous solutions, it is a strong reducing released from its proteins may induce further oxidative damage [26].
agent against some iron complexes. The reaction between Fe(III) and Although with the simplest representation, Fenton reaction seems as
O2•- is called as Haber-Weiss reaction, with the sequence: an outer-sphere electron transfer reaction, there are strong evi­
dences indicating that it occurs through an inner-sphere mechanism
Fe3+ + O2•− → Fe2+ + O2 including the formation of a transient L-Fe(H2O2)2+ complex. Here L
Fe2+ + H2O2 → Fe3+ + OH− + •OH can be a biological chelator (or phosphate or EDTA) [27,28].
In this case, the reaction steps can be shown as:
where the net reaction is:
2+
L-Fe + H2O2 → L-Fe(H2O2)2+
O2• + H2O2 → •OH + OH− + O2 [17]

2+
L-Fe(H2O2) → L-Fe3+ + •OH + OH- or
As can be seen from the above reaction steps, superoxide anion ra­
2+
dical is a potential reductant for Fe(III) ions, and the second step is a L-Fe(H2O2) → L-Fe4+ + 2OH- or
classical Fenton reaction. Haber-Weiss reaction is known as the source of L-Fe(H2O2)
2+
+ R → L-Fe3+ + OH- + ROH
generation of •OH by means of O2•− in biochemical systems. On the other
hand, the fact that the iron ions are not free but in the form of chelate R is the substrate and can be oxidized to ROH.
complexes may not prevent the reaction but even accelerate it, de­ After the formation of the L-Fe(H2O2)2+ transient species, it can
pending on the stability and coordinative saturation of the chelate [18]. yield different products, either Fe(III) or Fe(IV), through the reaction
HOO• is the protonated form of O2•-, and is stronger than su­ between ligand-bound Fe(II) and H2O2.
peroxide radical, both as an oxidant and reductant. The question
“how super is superoxide?” was asked by Sawyer [19] to indicate the 1.2. Antioxidants (AOXs)
moderate effectiveness of superoxide anion radical as a redox spe­
cies (i.e. both as an oxidizing and reducing agent). Since the pKa The AOX concept is a common interest for different disciplines
value of HOO• is 4.80, at physiological pH it easily turns into O2•- by such as bioanalytical chemists, physicians, dieticians and maybe
acidic dissociation. In the human body, superoxide dismutase group just for consumers. Therefore, the term AOX should be in its most
enzymes are responsible for consuming superoxide radicals [11]. basic form, simple and the same for all relevant groups. Probably
Superoxide can accept two protons and one electron to easily gen­ the most appropriate definition is that an AOX is a substance
erate H2O2. In the presence of certain oxidase enzymes, O2 can capable of impeding or retarding the unwanted oxidation of bio­
successively accept two electrons to form H2O2. The ground-state logically relevant substrates [29]. AOXs have the ability of reducing

3
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

oxidant species [2] usually by donation of electrons or H-atoms. developed by Benzie and Strain (1996), depends on the AOX oxi­
For food chemists, AOXs serve to prevent foods against deteriora­ dizing ability of ferric iron in the presence of 2,4,6-tripyridyl-s-
tion by means of inhibiting oxidation [30]. The use of AOXs to triazine (TPTZ) as ligand. Fe(III) can oxidize certain antioxidants in
prevent spoilage and prolong the lifespan of foods dates back to acidic medium, while itself being reduced to the blue-colored Fe(II)-
ancient times [31]. Apak mentioned two traditional Turkish foods TPTZ chelate, the absorbance of which is measured at 593 nm [38].
as examples of this phenomenon in his study. These are a soup The health beneficial effects of AOXs are usually associated with
named “tarhana” and herby cheese which have been known and their ability of removing reactive species. From this point of view,
made for over two hundred years. Both of them are made using biochemists, medicinal chemists and nutritionists often refer to sub­
certain herbs, vegetables and spices that are rich in phenolic AOX. stances that can quench metabolically produced (in vivo) reactive
The aim here is to prepare these foodstuffs in the summer and species as AOX. Therefore, these scientists are more focused on oxi­
store them during the winter when there are no fresh herbs and dative stress biomarkers in understanding health protection, and con­
vegetables, and consume them at this time [29]. Although the term sequently they are less concerned with TAC which is not exactly
AOX generally has positive connotations, there are some evidences correlated to disease prevention. On the other hand, some food sci­
to the contrary in the literature. Especially Halliwell and Gutter­ entists oriented at classifying and screening foodstuffs may understand
idge stated in their various studies that supplemental formulas AOXs as substances having high TAC and radical quenching ability in
containing AOX are not as beneficial as they thought in terms of vitro [39]. It can be concluded that although RS and TAC are the two
health [32,33]. Another important point is that in vitro and in vivo essential parts of redox biology, biological scientists are usually inter­
activities of some AOX compounds such as polyphenols can be very ested in RS assays while food chemists are interested in TAC assays.
different from each other. Studies on the effects of polyphenols There are a few studies in the literature aiming to fill this gap between
using cell culture indicate that they can act as prooxidants in these two types of assays. An example of this is the work of Çekiç et al.
certain conditions. Prooxidants may have both reducing and oxi­ The authors developed a method depending on the determination of
dizing character; they can induce oxidative stress, either by gen­ certain RS using N,N-dimethyl-p-phenylenediamine (DMPD) reagent,
erating reactive oxygen/nitrogen species (RONS) or by inhibiting where RS in the reaction medium caused the formation of pink colored
antioxidative defenses of the organism [34]. For example, under DMPD•+ radical cation from DMPD and the color intensity was directly
certain conditions, polyphenolic antioxidants may reduce transi­ proportional to the amount of RS. The observed color may be explained
tion metal ions to their lower oxidation states thereby triggering by an extended π-conjugation caused by a good electron donor (N atom
Fenton reactions giving rise to ROS. On the other hand, a mild of DMPD) and an acceptor (RS) system created by oxidation. However,
oxidative stress can be beneficial since it may trigger endogenous since these RS could be scavenged by AOXs, the color intensity de­
AOX defense system (e.g., by increasing GSH level). Therefore, if creased proportionally with AOX concentration. In this manner, both RS
there is no nutritional deficiency, excessive consumption of AOXs and TAC could be measured by using the same procedure [35]. This
should be avoided [25]. During the normal functioning of meta­ method, which was applied in a solution environment, was later
bolism, the prooxidant/antioxidant balance slightly favors proox­ turned into a sensor application by using a Nafion cation-exchange
idants to cause a mild oxidative stress and allows endogenous AOX membrane as a solid support material for capturing DMPD•+ [40]. Di­
(and repair systems) to interact to reduce disease incidence. It is rect determination of RS (especially very short-lived free radicals) is
still a controversial subject whether the use of synthetic AOXs is difficult and needs sophisticated and expensive instrumentation such
effective to prevent some diseases [11]. Foodstuffs such as fruits, as electron spin (paramagnetic) resonance (ESR/EPR). Therefore, in­
vegetables, grains and herbs, which are used as spices or beverages direct techniques based on the determination of products formed from
with a number of antioxidative ingredients (such as polyphenols, the reaction between RS and selected biological macromolecules (DNA,
tocopherols and carotenoids) usually serve better than only one proteins and phospholipids) are generally preferred. The RS causing
specific type of AOXs because of the collaborative action of AOXs. oxidative stress, specifically H2O2 and superoxide radicals, can be
Measuring plasma total antioxidant capacity (TAC) and its changes sensed by voltammetric and amperometric techniques [41], while RS-
may give useful hints to evaluate the health status of an individual induced oxidative conversion products on DNA and proteins can also be
person. Since the amount of AOX is directly proportional to redu­ measured by electrochemical techniques.
cing the rate of oxidants and/or suppressing (or delaying) oxida­ When RS cannot be counterbalanced by AOX molecules, a series
tion, there is usually an negative correlation between the of irreversible chemical modifications occur in the body. The pro­
concentration of TAC and total RS in the human body [35]. For ducts generated by means of these modifications may be considered
example, Bean et al. [36] successfully demonstrated this negative as “oxidative stress biomarkers”. Currently there are measurable
correlation between RS and TAC for bladder tissue undergoing biomarkers which are not clinically relevant yet [42]. Biologists re­
ischemia/reperfusion using the cupric ion reducing antioxidant searching free radicals have adapted this term from molecular epi­
capacity (CUPRAC) method. To briefly explain the CUPRAC method demiology [43], related to oxidation products generated from DNA,
introduced by Apak et al. [37], the copper(II)-neocuproine (Cu(II)- proteins and lipids and AOX consumption [44]. These biomarkers
Nc) reagent as the chromogenic oxidant can oxidize antioxidants, particularly include malondialdehyde (MDA) or isoprostanes for
while itself being reduced to the yellow-orange Cu(I)-Nc chelate. lipid oxidation; protein carbonyls for protein oxidation, and 8-hy­
The developed color is measured as absorbance at 450 nm against droxydeoxyguanosine (8-OHdG) for nucleic acid oxidation [45].
a reagent blank. The protons released during the reaction are
neutralized by an ammonium acetate buffer at pH 7 [37]: 2. Metabolic Oxidation Products
2+ + +
Cu(Nc)2 + AOX → Cu(Nc)2 + oxidized AOX + H
As a result of oxidative stress conditions, changes occur in the
structure and function of biomolecules such as DNA, lipids and
Bean et al. determined that while the level of free radicals in­ proteins [46], which will be disussed in this section.
creased, there was a decrease in protective AOX levels, measured by
two different assays [36]. The authors stated that they were able to 2.1. Lipid Peroxidation
demonstrate this negative correlation between RS and TAC using the
CUPRAC method [37], but the Ferric Reduction Antioxidant Power Lipids are essential components of cellular units such as plasma
assay (FRAP) [38] method, which was the other method they tried, membrane, Golgi apparatus, endoplasmic reticulum. Cell mem­
was not successful in this regard. The FRAP method, originally branes composed of lipid bilayers provide structural integrity by

4
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

Fig. 1. Chemical structures of linoleic and arachidonic acids.

surrounding the cell. The fluidity of a membrane significantly affects (cis-5,8,11,14,17-eicosapentaenoic). Linoleic acid and arachidonic
its functions. The presence of double bonds in lipids containing acid are shown in the Fig. 1 [51].
unsaturated fatty acids increases membrane fluidity. The more Lipid peroxidation takes place in three steps: initiation, propa­
double bonds a polyunsaturated fatty acid (PUFA) contains, the more gation and termination [52]. Lipid peroxidation can be initiated by
fluid it will be. At the same time, this property makes PUFAs sus­ removing a hydrogen atom from the methylene group of the PUFA
ceptible to autoxidation [47] based on the arrangement of the double side chain by a free radical. With the abstraction of a hydrogen atom
bonds within the molecule. Hydrogen abstraction is governed by the from the fatty acid, an unpaired electron remains on the carbon. The
bond dissociation energy (BDE) of the covalent C-H bond, which is unstable fatty acid radicals formed turn into conjugated diene
weakened by adjacent electron-rich –C]C– bonds in PUFAs that are structure in order to become thermodynamically more stable. In the
particularly open to H-atom abstraction due to the presence of single propagation step, fatty acid radicals also uptake oxygen to form the
or multiple CH2-interrupted hydrogens with lower BDEs. After the lipid peroxyl radicals which can start the chain reaction by ab­
formation of lipid radicals, the conjugated π-electrons of the double stracting hydrogen from another PUFA [53,54]. Generation of lipid
bond stabilize the free radical through delocalization over the diene peroxides from a PUFA was shown in Fig. 2.
structure and enable rapid reaction with triplet oxygen to form In the termination step, non-radical products such as alkanals
peroxyl radicals which can abstract another H-atom from the lipid and alkenals are formed. These compounds exhibit toxic properties
structure leading to hydroperoxide formation and free radical chain due to forming a Schiff base with amino groups, inactivating various
propagation [48]. enzymes, and interacting with thiol groups. As a result of lipid
In biological systems, lipid peroxidation can be induced by exo­ peroxidation in the cell membrane, the membrane transport system
genous sources such as air pollution, smoking, UV light or by free is affected, intracellular and extracellular ion balances are disrupted,
radicals generated by endogenous enzymatic systems such as membrane permeability changes [53].
NADPH oxidase and XO [49]. As a result of the peroxidation of The chain reactions can also terminate with the interaction of
phospholipids in the cell membrane with ROS, reactive lipid hy­ AOXs with lipid radicals. The AOX radical formed as a result of the
droperoxides and secondary carbonyl compounds such as 4-hy­ interaction of the AOX with the lipid peroxyl radical may turn into
droxy-2-nonenal, malondialdehyde, acrolein, crotonaldehyde, non-radical species by dimerizing or reacting with another lipid
methylglyoxal are formed [50]. peroxyl radical [55].
Lipids can undergo peroxidation, in particular, PUFAs with more Lipid peroxidation occurs through two different mechanisms:
than two cis-double bonds and their esters are easily oxidized by non-enzymatic peroxidation and enzymatic peroxidation [55]. Non-
free radical−induced chain oxidation. Major PUFAs are linolenic enzymatic peroxidation of lipids can be initiated with a radical as
acid (HOOC(CH2)7CH=CHCH2CH=CH(CH2)4CH3), docosahexaenoic described above or by generating radical species from non-radical
acid (cis-4,7,10,13,16,19-docosahexaenoic acid), arachidonic acid molecules [54]. In biological systems, this occurs through Fenton and
(cis-5,8,11,14-eicosatetraenoic acid) and eicosapentaenoic acid Haber-Weiss reactions in the presence of various transition metals,

Fig. 2. Steps of lipid peroxide generation from a polyunsaturated fatty acid (PUFA).

5
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

especially iron and copper [56]. Lipid peroxides can rapidly de­ IsoPs, initially bound to phospholipids, are freely found in bio­
compose in the presence of iron salts and form alkoxyl radicals and logical fluids after being released from cell membranes by phos­
peroxyl radicals [57]. pholipases [61,62]. The determination of IsoPs in biological fluids is
Enzymatic peroxidation of lipids occurs by enzymes such as li­ important in monitoring the treatment of diseases, as they are as­
poxygenases (LOXs), cyclooxygenases (COXs) and cytochrome P450 sociated with various diseases. Levels of F2-IsoPs, have been noted to
(CYPs). COXs, CYPs, LOXs are responsible for the synthesis of lipid be significantly increased in asthma [63], neurodegenerative dis­
endoperoxides, epoxyieicosatrienoic acids and lipid hydroperoxides, eases such as multiple sclerosis [64], AD [65,66] and cardiovascular
respectively [54]. disease [67].
Since docosahexaenoic acid (DHA) contains more double bonds
2.1.1. Lipid Peroxidation Products than AA, it is more susceptible to free radical-mediated peroxidation.
2.1.1.1. Isoprostanes. Isoprostanes (IsoPs) are widely used as Peroxidation of DHA, the most abundant fatty acid in the central
biomarkers in the assessment of lipid peroxidation in vivo due to nervous system, forms isoprostane-like compounds called neuro­
their stability, selectivity and specificity (Fig. 3). IsoPs are prostanes (NPs). The bicyclic endoperoxide intermediates formed by
prostaglandin-like molecules formed by the non-enzymatic the peroxidation of DHA are reduced to form F-ring NPs (F4-NPs)
peroxidation of arachidonic acid (AA). Various IsoP compounds are (Fig. 4) [58]. While AA is found in all cells, DHA is mostly found in
formed by oxidation of AA. Firstly, the arachidonyl radicals formed neuronal membranes. Therefore, the determination of F4-NPs
by the peroxidation of AA undergo endocyclization to produce four formed by the oxidation of DHA allows the determination of oxi­
positional bicyclic endoperoxide intermediate isomers, known as dative damage in neuronal membranes specifically [68].
5-, 12-, 8-, or 15-series, according to the carbon to which the Additionaly, as oxygen tension increases, isofurans (IsoFs) and
hydroperoxyl in the side chain is attached. These intermediates are neurofurans (NFs) can be formed, respectively, by the peroxidation
then reduced to form F2-isoprostan (F2-IsoP) regioisomers [58–60]. of AA and DHA. The plausible mechanisms for the formation of NFs

Fig. 3. The oxidation and peroxidation products of arachidonic acid.

6
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

Fig. 4. DHA oxidation products.

include cleavage of cyclic peroxides and hydrolysis of epoxides mediated oxidation results in hydroperoxyoctadecadienoic acids
[69,70]. The formation mechanism of these compounds occurs si­ (HPODEs). There are four HPODE isomers (Fig. 5) 13-hydroperoxy-
milarly to F2-IsoPs [58]. 9(Z), 11(E)-octadecadienoic acid (13-(Z,E)-HPODE); 13-hydroperoxy-
9(E), 11(E)-octadecadienoic acid (13-(E,E)-HPODE); 9-hydroperoxy-
2.1.1.2. Lipid Hydroperoxides and Lipid Hydroxides. Hydroperoxides 10(E), 12(Z)-octadecadienoic acid (9-(E,Z)-HPODE) and 9-
are primary products formed as a result of peroxidation of PUFA and hydroperoxy-10(E), 12(E)-octadecadienoic acid (9-(E,E)-HPODE) [62].
cholesterol (Ch) by lipoxygenase or free radical mediated peroxidation. Hydroperoxides formed as a result of the peroxidation of PUFAs are
Linoleates are the most abundant PUFAs in vivo, and their radical- reduced by glutathione peroxidases and selenoprotein P to their

Fig. 5. Hydroperoxyoctadecadienoic acid (HPODE) isomers.

7
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

corresponding hydroxides and form reactive secondary degradation (dCyd) adducts, and these DNA-MDA adducts have mutagenic effects
products [51]. in bacteria and mammalian cells [72].
Some reactive secondary degradation products are formed by the Phospholipids containing linoleic acid and AA are considered the
decomposition of lipid hydroperoxides, which are the primary pro­ main source for 4-HNE production [75]. 4-hydroxy-2-nonenal and 4-
ducts of the peroxidation of PUFAs. Hydroxy acids are formed by the hydroxy-2-hexenal (HHE) are formed as a result of peroxidation of
reduction of lipid peroxides. Lipid hydroxides can be produced by ω6 and ω3 fatty acids, respectively. 4-hydroxy-2-alkenals are highly
reduction of hydroperoxides, and can also be formed by enzymatic reactive due to their three functional groups (carbonyl, -C = C-
peroxidation, singlet oxygen peroxidation, or radical mediated per­ double bond and secondary alcohol) they have, and under the in­
oxidation. While cis-trans hydroxides are formed by all three per­ fluence of these groups they can interact with biomolecules con­
oxidation pathways, trans-trans hydroxides are formed only as a taining amino and thiol groups through Michael additions and Schiff
result of free radical mediated peroxidation; thus, the latter serve as base formation [71]. In addition to biochemical modifications, it may
specific biomarkers of free radical induced lipid peroxidation [51]. cause changes in the biophysical properties of membranes, such as
Due to their relative chemical stability and consistency of measured membrane fluidity, depending on its concentration [76].
levels in human fluids and tissues, lipid hydroxides may be con­ Lipid peroxidation products such as 4-HNE and MDA can be
sidered to be more suitable as biomarkers than hydroperoxides [51]. found in free form in metabolism; they can also be covalently bound
Hydroperoxyeicosatetraenoic acid (HPETE), which is formed by the to various molecules such as proteins and nucleic acids due to their
peroxidation of AA, and hydroperoxyoctadecadienoic acid (HPODE), high reactivity [46]. It has been stated that the formation of these
which is the peroxidation product of linoleic acid, can be reduced to products is associated with inflammation, apoptosis, and various
hydroxyeicosatetraenoic acids (HETEs) and hydroxyoctadecadienoic degenerative diseases [77]. Acrolein, one of the highly reactive
acids, respectively, with a strong reducing agent [54]. products of the metal catalyzed oxidation of PUFAs, can also induce
neuronal damage by forming addition products with proteins [68].
2.1.1.3. Aldehydes. As a result of lipid peroxidation, PUFAs can form
various short chain aldehydes, including 2-alkenals, 4-hydroxy-2- 2.1.1.4. Oxysterols. Ch is a sterol lipid found in the lipoproteins and
alkenals and ketoaldehydes (Fig. 6) [58]. These are secondary membranes of eukaryotic cells. Since Ch is a monounsaturated lipid,
metabolites of lipid peroxidation. Aldehydes are the most common it is less sensitive to peroxidation than substances containing
degradation products of lipid peroxides [54]. The main ones are 4- polyunsaturated fatty acids. Peroxidation of Ch can occur in
hydroxynonenal (4-HNE) and MDA. enzymatic or radical-mediated modes. Peroxidation in the cyclic
MDA may be formed as a result of enzymatic and free radical part of Ch is generally radical-mediated, while peroxidation in the
peroxidation of PUFAs containing at least three double bonds [71]. In side chain is usually enzymatic [78]. Ch peroxidation products used
addition, MDA is formed in prostaglandin synthesis [72]. MDA is as biomarkers of oxidative stress are called oxysterols. These
frequently used as a lipid peroxidation marker. MDA can mainly oxysterols (Ch oxides) include hydroperoxides, diols, epoxides and
react with lysine (Lys) residues of proteins to form adducts with ketones. As a result of the peroxidation of Ch, different oxidation
proteins and DNA. It plays a role in the pathogenesis of various product such as 7α- and 7β-hydroperoxycholesterol (7α-OOHCh and
diseases such as diabetes mellitus [73] and neurodegenerative dis­ 7β-OOHCh), 7α- and 7β-hydroxycholesterol (7α-OHCh, 7β-OHCh), 5α,
eases [74]. It has been reported that MDA reacts with DNA bases to 6α- and 5β, 6β-epoxycholesterol and 7-ketocholesterol (7-KCh) are
form deoxyguanosine (dG), deoxyadenosine (dA) and deoxycytidine formed (Fig. 7) [62]. Ch can also be oxidized by enzymes, most of

Fig. 6. Secondary oxidation products of lipids.

8
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

Fig. 7. Cholesterol peroxidation products.

which belong to the cytochrome P450 class. 27-hydroxycholesterol, As a result of oxidative damage of proteins, some changes may
which is an intermediate product in bile synthesis from CYP27A1 occur in their physical or chemical properties such as solubility,
enzyme in the inner membrane of mitochondria, 24(S)- structural integrity, and enzyme activity [81]. It has been reported
hydroxycholesterol is produced from CYP46 enzyme, especially that the levels of protein oxidation products increase in various
found in neurons, and 7α-hydroxycholesterol is produced from diseases related to aging such as AD [85], diabetes [86], and catar­
CYP7A1 enzyme. 25-hydroxycholesterol and 7α-hydroxycholesterol acts [87].
can be formed by both enzymatic and radical mediated
peroxidation [79]. 2.2.1. Oxidation of the Protein Backbone
In the first stage, as a result of the interaction of the amino acid
with the radical, a hydrogen atom is removed from the α-carbon, and
2.2. Protein Damage a carbon-centered radical is formed. Subsequently, an alkylperoxy
radical is formed by the addition of O2. At this stage, the carbon
Proteins become an important target for oxidizing agents due to centered radical can react with another carbon centered radical in
their abundance in cells, plasma and tissues and their high reaction the absence of oxygen and form protein-protein crosslinks. As a
rate with various oxidants [80]. The interaction of proteins with result of the reaction of the alkyl peroxy radical with HO2• or Fe2+
reactive oxygen species can result in oxidation of amino acid side and H+, alkyl peroxide is formed. The hydrolysis of the imino deri­
chains or the protein backbone, the formation of carbonyl-derived vative, which is formed by the decomposition of the alkyl peroxide,
compounds, and protein-protein crosslinking [81]. Since proteins forms an aldehyde or an α-keto acid [88].
can contain different amino acids in their side chains, a large number
of radicals can occur on proteins. Amino acids with a thiol group and 2.2.2. Oxidation of Amino Acids
aromatic side chain, namely cysteine and tyrosine, respectively, are Cysteine and methionine are very sensitive to oxidative mod­
the most sensitive moieties to oxidation [82,83]. ification due to the nucleophilic character of the thiol and thiol ether
In metal-catalyzed protein oxidation, transition metal ions such groups, respectively, that they contain. The oxidation of cysteine
as Fe (II) and Cu (I) bind to the metal binding site in proteins and residues begins with the formation of sulfenic acid and sulfinic acid
then react with H2O2 to form •OH radical that attacks amino acid and can continue with the formation of sulfonic acid, an irreversible
residues near this site [84]. further oxidation product. The oxidation of cysteine to sulfenic and

9
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

sulfinic acid derivatives is reversible [89]. Since these species are Similarly, 8-hydroxyadenine (8-OH-Ade) and 4,6-diamino-5-for­
unstable, they can form oxyacids by hydrolysis, and can also form mamidopyrimidine (FapyA) are formed from adenine. By radical-
disulfide bonds by reacting with another thiol group or in the pre­ induced modification of thymine, compounds such as thymine
sence of a donor such as glutathione [81]. Cystine, in which the two glycol, 5-(hydroxymethyl) uracil, and 5-formyluracil (5-FoU) are
cysteines are linked by disulfide bonds, is an important cysteine formed. Some modifications of the cytosine base are cytosine glycol,
modification. The formation of these disulfide bonds is effective in 5-hydroxycytosine (5-OHC) and 5-hydroxyuracil (5-OHU). The re­
stabilizing proteins. lated DNA oxidation products are shown in Fig. 8 [96].
As a result of the oxidation of methionine residues in the side The hydroxyl radical reacts with the sugar portion of DNA to
chains of proteins, a mixture of stereoisomers of methionine sulf­ generate carbon-centered sugar radicals, which results in breaking
oxide is formed. Methionine sulfoxide can be reduced back to me­ the DNA strand, forming various sugar products and abasic sites (i.e.
thionine by the methionine sulfoxide reductase enzyme [90]. As the apurinic/apyrimidinic sites as common lesions in DNA) [99]. DNA-
further oxidation product of methionine sulfoxide, methionine sul­ protein cross-linking can occur by adding the DNA base radical
fone is formed. However, this product cannot be reduced again by (formed by the interaction of DNA with radicals) to the aromatic
methionine sulfoxide reductases [89]. amino acid of proteins or amino acid radical [100].
In addition, the tyrosyl radical formed by the oxidation of tyr­
osine side chains can form dityrosine by dimerizing with another
3. Antioxidants
tyrosyl radical, or it can also form 3,4-dihydroxyphenylalanine by
interacting with hydroxyl radicals [89]. As a result of the oxidation of
Considering that, free radicals, produced in the body, may cause
proteins; nitrotryptophan or kynurenine from tryptophan residues,
oxidative damage on biomacromolecules such as lipids, proteins and
2-oxohistidine from histidine residues, 3-, 4-, 5- hydroxyleucine
DNA, the importance of AOXs that delay or prevent this damage by
derivatives from leucine residues can be formed. Some amino acid
scavenging reactive species cannot be underestimated. AOXs can
residues such as Lys and arginine also turn into various carbonyl
have different functions depending on the type of radical species
derivatives [90,91].
produced and of the target molecule protected [101]. As AOXs can
reduce the risk of degenerative disease development by counter­
2.2.3. Protein Carbonyl Derivatives
balancing an excess of reactive species, they are vital for human
The carbonylation of proteins is irreversible and can cause pro­
health [102]. Furthermore, AOX intake from exogenous sources is
teins to lose their activity since it causes a change in the peptide
very important as oxidants in the body may increase in various
chain structure [92]. Carbonyl derivatives are frequently used as
disease conditions. While some AOXs can be naturally synthesized in
biomarkers in the oxidative damage of proteins and can be formed
the body, some are taken up through nutrition. Among the various
mainly in four different ways. The first of these is the direct oxida­
classes of AOXs, enzymatic AOXs such as SOD, CAT and glutathione
tion of proline, arginine, Lys and threonine residues in protein side
peroxidase; hydrophilic ones such as urate, ascorbate, glutathione
chains. As a result of this, various carbonyl compounds such as γ-
and flavonoids; lipophilic ones such as tocopherols, carotenoids and
glutamyl semialdehyde, α-aminoadipic semialdehyde, 2-amino-3-
ubiquinol can be mentioned [103]. Aside from their health-beneficial
ketobutyric acid are formed [81]. Also, they can be produced by
effects, AOXs in foods, food supplements and pharmaceuticals pro­
cleavage of protein backbones by the α-amidation pathway or oxi­
long their shelf life. AOXs can be classified as primary or secondary
dation of glutamyl residues. Oxidation by α-amidation pathway re­
AOXs depending on their mechanism of action. Primary AOXs sca­
sults in N-α-ketoacyl derivative compounds [89]. Furthermore,
venge radical species by H-atom or electron transfer, while sec­
carbonyl derivative compounds are formed by the Michael addition
ondary AOXs inactivate prooxidant catalysts. Although synthetic
to cysteine, histidine and Lys residues of aldehydes, which are sec­
AOXs are known as primary AOXs, it has been reported that they can
ondary oxidation products of lipids, or by the reaction of Lys residues
be toxic and carcinogenic depending on their concentrations [102].
with reducing sugars or their oxidation products such as ketoamines
AOXs are also divided into three subgroups according to their de­
and ketoaldehydes [81,92]. The products formed as a result of a
fense mechanisms: (i) preventive, (ii) radical scavenger and (iii)
series of reactions of Lys residues with the carbonyl groups of car­
enzyme repairing. Plant-based AOXs are generally hydrophilic,
bohydrates called glycoxidation are advanced glycation end products
however tocopherols, vitamin K and carotenoids are hydrophobic.
and are used as biomarkers in the oxidative damage of proteins [93].
Uric acid and ascorbic acid are water-soluble AOXs [104]. The con­
jugate base (salt) form of phenolic AOXs is more soluble in the
2.3. DNA Damage
aqueous phase than the parent free acid (at pH above pKa of
acid-form).
The interaction of DNA with reactive oxygen species can cause
DNA-protein cross-linking, the formation of sugar and base mod­
ification products, intra-strand and inter-strand cross-linking, single 3.1. Enzymatic Antioxidants
or double strand-breaks [94]. Damage to DNA caused by ROS origi­
nating from endogenous or exogenous sources may increase the risk SOD, CAT, GPx are the best-known AOX enzymes. Since they have
of developing various diseases such as cancer, cardiovascular dis­ high affinity for ROS, they provide more effective protection in case
eases, and neurodegenerative diseases [95]. of oxidative stress [103].
A large number of sugar and base modification products have Coenzyme Q10 is a lipid-soluble AOX and, apart from its radical
been described, depending on the reaction conditions [96]. Of the scavenging effect, also plays a role in the regeneration of various
DNA bases, guanine is sensitive to oxidation due to its lowest redox vitamins A (retinol), C (ascorbic acid) and E, and in metabolic events
potential [97] and highest oxidizability; it can be easily oxidized to such as inhibiting the degradation of biomacromolecules such as
8-oxo-7,8-dihydrodeoxyguanine via single-electron transfer to ROS lipids, DNA and proteins [105].
such as H2O2, superoxide and hydroxyl radicals [98]. DNA mod­ SOD, which is a metalloenzyme, is one the most effective enzy­
ification causes various mutations, mainly GC-TA transversion matic AOXs found in all subcellular compartments and acts as a
mutation, due to mispair [42]. Two important products resulting protective agent against the toxic effects of high amounts of ROS
from the guanine modification are 8-hydroxyguanosine (8-OH-G) [106]. SOD catalyzes the breakdown of superoxide into oxygen and
in the presence of oxygen and 2,6-diamino-4-hydroxy-5-for­ hydrogen peroxide. According to their metal cofactors, there are
mamidopyrimidine (FapyG) in the presence of reducing agents. three types SOD enzymes in the human body: cytosolic CuZn-SOD,

10
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

Fig. 8. The main oxidation products of DNA bases.

mitochondrial Mn-SOD and Fe-SOD [103,106]. It helps prevent the


transition metal-catalyzed formation of hydroxyl radicals [106].
Catalases are enzymes with tetrameric heme group and are in­
volved in the detoxification of H2O2 produced in peroxisomes by
oxidases involved in various catabolic events [106]. It catalyzes the
decomposition reaction of hydrogen peroxide into the nontoxic
products of water and oxygen (via the reaction: 2 H2O2 → 2 H2O +
O2) [107]. This enzyme, which can break down about 6 million H2O2
molecules per minute, has almost the highest turnover rate [106].
In addition, CAT has also been reported to be effective on some
toxic compounds via peroxidative reaction [103]. Catalases (hydro­
peroxidases) have been studied extensively to date, containing more
than 300 types, namely monofunctional catalases, bifunctional
catalases and manganese containing catalases [107]. Fig. 9. Glutathione reductase and involved cellular redox cycles (GSH: reduced glu­
On the other hand, transition metals such as Fe, Cu and Mn in tathione, GSSG: oxidized glutathione, ASA: ascorbate, NAD(P)H: nicotinamide adenine
dinucleotide phosphate.
different oxidation states can act both as prooxidants and AOXs.
These metals can transfer their electrons to form radicals and cause
oxidative stress. At the same time, since these metals constitute the decomposes hydrogen peroxide into water. Especially when free or
active redox centres of AOX enzymes, they play an important role in weakly-complexed Fe and Cu ions are present in excess, they can
the biological AOX defense system. As being the active centres of cause peroxidation and act as prooxidants through Fenton and
metalloenzymes, Cu, Zn and Mn in superoxide dismutase convert Haber-Weiss reactions, but metals that are more tightly bound to
superoxide anion to hydrogen peroxide, while Fe in catalase protein structures such as ferritin prevent these structures from

11
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

Fig. 10. Chemical structure of β-carotene.

Fig. 11. Chemical structure of α-tocopherol.

acting as prooxidants [108]. It should be noted that although Fe in photosynthetic bacteria [112]. Carotenoids with strong AOX prop­
ferritin is relatively tightly bound, its release under severe condi­ erties are divided into two main groups: (i) carotenes (ii) xantho­
tions (such as redox-mediated chelation) is possible. There are stu­ phylls. These two groups differ from each other in terms of their
dies in the literature indicating that certain agents such as chemical structures. While the first group contains carbon and hy­
superoxide anion can promote iron release [109]. drogen in its structure, the second group compounds contain at least
Glutathione reductase, which is commonly found in chloroplasts, one oxygen atom [113]. This group contains lipid-soluble AOXs, and
plays an important role in the ascorbate-glutathione cycle. This its another important role is to form vitamin A by enzymatic clea­
enzyme catalyzes the reduction reaction of glutathione, a low-mo­ vage in the body [110]. There are more than 600 carotenoids in
lecular-mass AOX, and plays a role in many metabolic events. As can nature, of which the most abundant is β-carotene found in orange
be seen from Fig. 9, glutathione reductase not only plays a role in the and yellow fruits, and green leafy vegetables. Studies have also
reduction of glutathione, but also protects the ascorbate-glutathione proven that carotenoids can scavenge 1O2 [106,114]. There have been
cycle and acts as a ROS scavenger [106]. many studies on β-carotene, lycopene, lutein, and zeaxanthin and
The glutathione-S-transferase enzyme mainly catalyzes the re­ their protective effects, especially against cancer and eye diseases
action of tripeptide glutathione with electrophilic xenobiotic sub­ [115]. Zeaxanthin and lutein are the main components of the eye-
strates. In addition, it is involved in the conversion of toxic protecting macular pigments [116]. Foods rich in carotenoids are
substances such as hydroxyperoxide and pesticides into nontoxic usually colorful, and the deterioration in color is associated with
metabolites in hormone homeostasis [106]. reduced or oxidized forms of these carotenoids [117]. Their AOX
activity show differences according to the location of double bonds
3.2. Non-Enzymatic Antioxidants in the molecule, their coexistence with other AOXs, and their con­
centration. Similar to most other AOXs, they can act as prooxidant
Vitamin E, carotenoids, polyphenols and ascorbic acid are non- when they are in excessive amounts [115].
enzymatic AOXs that have important physiological functions, and Another compound that is an AOX precursor, such as carotenoids,
since they cannot be synthesized in the body, they should be sup­ is glutamine and is particularly beneficial for the liver, intestinal
plied by nutrition. Vitamin E exists in eight different forms, four functions, and the immune system [115]. N-Acetyl cysteine, the
tocopherols and four tocotrienols, the essentially used form of which source of sulfhydryl, is a thiol compound that acts as a radical sca­
is α-tocopherol. Coenzyme Q10 (CoQ10) or ubiquinone is found in venger [105]. Also, N-Acetyl Cysteine, which is a glutathione pre­
the biological system which is synthesized by the conjugation of cursor, can increase its AOX effect by increasing the glutathione
benzoquinone ring with an isoprenoid chain, but it can be supple­ level. It has also been used as a criterion to investigate the effect of
mented through diet to further energize the body through ATP ROS on various diseases [115].
production. Nutrition and diet are an important part of the AOX α-Lipoic acid is a low-molecular-mass cyclic disulfide and soluble in
defense mechanism [103]. Glutathione, uric acid and bilirubin are both lipid and water, and takes part in various metabolic events such as
also small molecular mass AOX present in human fluids [110]. Vi­ regulating glucose metabolism and the circulation of various anti­
tamins C and E are known as AOX vitamins, together with β-carotene oxidants in blood [105]. In the human body, lipoic acid is reduced to
(actually a provitamin A carotenoid) (Fig. 10), and play an important dihydrolipoic acid by means of lipomide dehydrogenase enzyme. It is
role in preventing some diseases caused by oxidative damage [111]. known that this reduction may be completed with the aid of other
Carotenoids are an important group found in fruits and vege­ cellular reducing systems [118]. Lipoate may also enhance the favorable
tables that can prevent cancer due to their AOX functions [110], and effects of other AOX systems such as SOD, CoQ10 and glutathione, and
they are natural pigments which synthesized by plants, algae and may regenerate the oxidation state of vitamins C and E.

12
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

Glycine is another non-enzymatic AOX precursor amino acid that AOX, because it can inhibit lipid peroxidation by complexing the Cu
takes part in the synthesis of glutathione and proteins acting as (II) ion. Nicotinamide also acts as an AOX in preventing lipid per­
AOXs, and is especially involved in protecting the kidneys. Studies oxidation caused by photosensitization [115].
have also showed that glycine has a protective effect against The bioavailability of non-enzymatic dietary AOXs depends on
ROS [115]. many factors such as food processing, AOX structure and its lipo­
At high concentrations, vitamins C and E can act as prooxidants philic-hydrophilic balance, solubility and stability, stabilizing effect
[108]. There are 8 types of vitamin E; α, β, γ, and δ-tocopherol; α, β, γ, of food matrix, etc. [103]. After it was reported that synthetic AOXs
and δ-tocotrienol. α-tocopherol is the most effective one among may have toxic effects, studies were focused on phenolic com­
them (Fig. 11). The reason for this is the -OH group attached to pounds, which are sources of natural AOXs [102].
carbon number 6 in its structure [119]. The H-atom of the -OH group The AOX properties of phenolic compounds, which constitute a
in the chromanol ring can be given to a free radical, giving rise to a large part of the non-enzymatic AOX class, depend on their struc­
delocalized and stabilized unpaired electron; the resulting vitamin E ture. Phenolic compounds have aromatic ring structures containing
radical is kinetically inert so as to terminate the chain reaction of at least one OH group. Simple phenols, phenolic acids, acet­
lipid peroxidation [120] Vitamin E is a fat-soluble and chain- ophenones and phenyl acetic acids, hydroxycinnamic acids, hydro­
breaking AOX owing to its reaction ability against lipid peroxide xyanthraquinones, stilbens, flavonoids, lignans, biflavonoids and
radicals, and prevents this radical from reacting with a new PUFA. tannins are the main phenolic structures [123]. Different structures
Vitamin E radical, which takes part in the defense against lipid of flavonoids such as flavonols, flavones, flavanols, flavanones, an­
peroxidation, is converted into vitamin E by vitamin C at the water- thocyanidins and isoflavonoids are formed with the differences in
lipid interface [108], enabling the regeneration of vitamin E by vi­ the C ring (Figs. 12 and 13) [124]. Plant polyphenols have anti­
tamin C via favourable redox potentials. Another important role of bacterial, antiviral, antitumor and AOX properties. Polyphenols are
vitamin E is to prevent the formation of N-nitroso compounds in the multifunctional, and can be classified as primary AOXs found as
stomach, thus having a protective effect against various diseases electron donors, H-atom donors and radical scavengers. The ability
[110]. So, Vitamin E can both act as a direct radical scavenger and as to chelate metal ions is another important feature of polyphenols, as
an AOX by regulating various enzymes such as SOD, CAT, GPx [115]. the presence of transition metal ions catalyzes reactions that can
Ascorbic acid is a water soluble AOX widely available in plants; it initiate oxidative degradation, such as the Fenton reaction [125].
is a highly effective radical scavenger and reducing agent in aqueous Flavonoids are taken through the daily diet, and onions, cab­
medium [106,108]. One of the important tasks of vitamin C is to bage, broccoli, parsley, citrus fruits, grapes and legumes are accepted
prevent the formation of N-nitroso compounds in the stomach and as good sources. Due to its AOX properties, they prevent lipid per­
support the formation of collagen. For this reason, it has a protective oxidation and have protective effects against oxygen
effect against various diseases, including cancer [110]. Vitamin C can radicals. Considering that oxygen radicals have important effects on
also act as a direct ROS scavenger as well as recycler of α-tocopherol, atherosclerosis, cancer and chronic inflammation, the importance of
which plays an important role in lipid peroxidation. In addition, it is flavonoids taken through food becomes clear once again [127]. It has
important for the prevention of neurodegenerative diseases due to been reported that AOXs taken through diet, especially in the form of
its role in the transport of drugs that cannot cross the blood-brain fruits and vegetables, have a protective effect against many diseases.
barrier [115]. For example, Qui et al. worked on the design and Although this protective effect is mostly attributed to the vitamin C
synthesis of ibuprofen prodrugs combined with ascorbic acid [121]. and β-carotene content, plant-derived phenolics such as flavonoids
In addition, it was stated in literature that the conjugation of lipo­ also have an important role [124].
some ligands with ascorbic acid (i.e. ascorbic acid thiamine disulfide The chemical structure of flavonoids depends on the degree of
delivery system) may be an effective strategy to enhance central hydroxylation, presence of other substitutions, distribution of con­
nervous system drugs’ delivery ability into brain [122]. jugated double bonds, and the structural class in which they are
GSH is a water-soluble, low-molecular-mass antioxidant. GSH found. The hydroxyl group or glycosylation of the C3 group in the C
enables the reduction of hydroperoxides by some hydroperoxins and ring can change the photostability of the molecule. As mentioned
GPx, thus playing an important role in the cell defense system. At the before, changes in A, B and C rings in the flavonoid structure create
end of the reaction, some reactions catalyzed by glutathione-S- the structure of different classes. The biological activities of flavo­
transferase are observed and GSH is oxidized to a GSSG form. GSH noids and their metabolites are mainly affected by changes in this
can be reversibly oxidized to GSSG (glutathione disulfide) by the chemical structure. Quercetin having an optimal combination of the
action of oxidants and free radicals. There is a delicate balance be­ listed AOX properties is one of the most abundant flavonols in fla­
tween the sulfhydryl and disulfide forms of glutathione, where they vonoid-rich food plants [128].
play a protective role in cellular redox homeostasis called ‘dynamic The AOX mechanisms of flavonoids mainly proceed in three dif­
thiol/disulfide homeostasis’, and the ratio of reduced-to-oxidized ferent ways: Radical scavenging effect by H-atom transfer or electron
forms of glutathione (GSH/GSSG) is considered as a well-being in­ donation, prevention of metal-catalyzed formation of reactive spe­
dicator important for human health. Redox homeostasis is generally cies via metal chelating effect, and interaction with other
understood as a dynamic equilibrium between electrophiles and AOXs [123].
nucleophiles to maintain an optimal redox steady state [120]. In Phenolic compounds can be distributed differently in plant fa­
general, antioxidant biothiols may react with free radicals while milies. Flavonols, and to a lesser extent, flavones are found in most
being oxidized to thiyl radicals (RS•) and show a protective effect by plants along with hydroxycinnamic acid derivatives. Flavanones and
preventing the target cell from being damaged, the effect being flavones are found together in most plants and connected by en­
dependent on the reactivity of the RS• formed [115]. zymes. However, most plants rich in flavanone have almost no an­
Proline is known as an inhibitor of programmed cell death (PCD). thocyanins [124].
This is why it is referred to as a powerful AOX that reduces oxidative Some phenolic compounds such as butylated hydroxyanisol
stress. In addition, it has been proven in scientific studies that pro­ (BHA), butylated hydroxytoluene (BHT), tert-butylhydroquinone
line is a hydroxyl radical scavenger [106]. (TBHQ), 2-tert-butyl-4-methylphenol (TBMP) are called synthetic
Histidine is both a singlet oxygen (1O2) scavenger and a chelator AOXs which are used as food additives. These synthetic AOXs
capable of complexing Cu(II) ions. It is an amino acid that acts as an transfer their hydrogen atoms to lipid radicals to prevent the radical

13
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

Fig. 12. Chemical structures of flavonoids.

chain reaction involved in lipid peroxidation, as shown with the has side effects up to DNA damage when taken at high doses. BHA,
reaction equations: which prevents radical formation and lipid oxidation, is a heat-
resistant synthetic AOX used as a preservative in dry cereals,
ROO• + ArOH→ ROOH + ArO•
mixtures or desserts, and especially in fried products. It has been
ROO• + RH → ROOH + R• (+O2→ ROO•) reported that it can be a tumor initiator if taken at high doses.
TBMP has been used as an intermediate of AOXs and UV stabilizers
TBHQ, which is a synthetic AOX used in some foods, is espe­ used to reduce oxidative processes. Propyl gallate (PG), which is as
cially useful for the stabilization of unsaturated oils. However, it effective as vitamin E, plays a protective role in lipid peroxidation.

14
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

significant proportion of serum TAC. Serum or plasma TAC con­


centrations may reflect both RS overload and the level of dietary AOX
intake [130]. Oxidative stress biomarkers can be used as beneficial
tools to assess in vivo AOX activity/capacity, Apak et al. mentioned
this concept in their comprehensive review on AOX activity/capacity
measurement [133]. The most vulnerable targets for RS in the
human body (namely lipids, proteins and DNA) can be used as rea­
listic probe materials for determining the activity or capacity of AOX
substances. Because AOX can prevent or delay the oxidation of these
probes, the degree of this inhibition (or delay) can be used to eval­
uate the efficacy of the AOX compounds tested. For example,
Fig. 13. Stereochemistry of flavanones (naringenin). Uzunboy et al. used DNA as probe material. The authors created
oxidative damage on DNA by means of radical species obtained by
Fenton-type reactions and were able to determine the oxidative
It acts as a preservative in foods, especially in polyunsaturated damage products with the CUPRAC method, which they modified in
fats [129]. accordance with the purpose. They also added AOX compounds to
the reaction medium during radical formation and measured the
4. Analytical Strategies for the Measurement of Oxidative Stress decrement in the formation of oxidative damage products, which is
and Related Antioxidant Defense directly related to the concentrations of AOX compounds in the
system [134,135].
As mentioned earlier, both RS and AOX can be beneficial or
harmful depending on certain conditions, and the balance between 4.1. Determination of Lipid Oxidation
these two types of compounds is crucial in the human body.
Although there are different methodologies for detection of RS in Lipid peroxidation is a radical chain reaction consisting of three
vitro or in animal studies, to apply these strategies to patients is not basic steps: I- initiation (formation of lipid radical, L•); II- propaga­
easy. Therefore, oxidative stress status is usually determined by tion (producing lipid peroxyl radicals through oxygen uptake by lipid
measuring the products (biomarkers) produced by the oxidation of radicals); this can result in the formation of new lipid radicals; III-
certain molecules in biological samples such as blood or urine inhibition and termination by formation of nonradical products or of
[42,50]. The most well-known biomarkers used to detect oxidative much less reactive radicals. Especially in the presence of chain-
stress are MDA, protein carbonyls and 8-OHdG, which are terminal braking AOX compounds via the reaction between lipid oxidizing
products of oxidation reactions. At this point, it can be argued radicals and AOX, the more stable AOX radicals (such as ArO•) gen­
whether we measure “oxidative stress” or “oxidative damage”. erate [136].
Ghezzi classified oxidative stress biomarkers into four subgroups. The initial product of lipid peroxidation is peroxyl radical, which
According to this classification, the oxidative stress biomarkers can be detected directly by using ESR technique. In this method, the
showing oxidative damage of lipid, proteins and nucleic acids are spin signal of unpaired electron is used, but since this radical species
collected as ‘type 1’ biomarkers. The molecules that decrease at the is very unstable (transient), a radical spin trap should be used [137].
end of the oxidation with RS (e.g., glutathione (GSH)) are also in­ However, ESR is not a highly sensitive technique and needs steady-
cluded in type 1. It should be mentioned here that although the ratio state radicals in the high micromolar concentration range [138].
of reduced-to-oxidized forms of glutathione (GSH/GSSG ratio) is not Although ESR can be applied in vitro, it is not suitable for use in vivo.
an exact oxidative stress biomarker (because of the reversibility of One of the reasons for this phenomenon is the need to use spin
thiol oxidation to disulfide), it reflects cellular redox homeostasis trapping agents such as 5,5-dimethyl-1-pyrroline N-oxide (DMPO),
and is usually accepted as an indicator of well-being. On the other which have high levels of unknown toxicity in vivo. The other pos­
hand, there are certain enzymes in the human body that are able to sible reason is the scavenging of DMPO-radical adducts by the en­
produce RS like H2O2. Molecules that activate RS-generating en­ zymatic metabolism and endogenous AOXs such as ascorbate [139].
zymes are accepted as ‘type 2’ biomarkers. Uric acid (UA) and al­ Among the main indicators of oxidative damage on lipids, ethane
lantoin, which are stable products generated by means of XO and pentane in exhaled gas, lipid hydroperoxides, conjugated dienes,
enzyme, are type 2 biomarkers. Hypochlorous acid (HOCl), which and isoprostanes (i.e. prostaglandin isomers generated in vivo from
shows phagocytic H2O2 production by means of myeloperoxidase, is polyunsaturated fatty acids via a free radical induced mechanism)
another type 2 biomarker. The enzymes and small molecular mass can be mentioned [140].
biological AOXs such as vitamin C, E and bilirubin can scavenge RS in
the human body. They are defined as ‘type 3’ biomarkers and the 4.1.1. Determination of Lipid Hydroperoxides
levels of RS producing enzymes such as NADPH oxidases can also be According to the reaction between iodide (I-) and hydroperoxides
measured within this type of biomarkers. And finally ‘type 4’ bio­ in acidic medium, I2 (or its aqueous-soluble form, triodide: I3-) is
markers include the degree of mutations of different enzymes re­ liberated, which can be determined spectrophotometrically or ti­
sponsible for RS formation or scavenging which may indicate the trimetrically. Spectrophotometric determination of I3- is conducted
role of oxidative stress in a certain disease [45]. at 360 nm by measuring the absorbance of triiodide complex ion
There are different analytical strategies for in vitro determination formed from I2 and excessive I- [141]. For higher concentrations, the
of oxidative stress. They can be classified according to the bioma­ very well-known thiosulfate titration in the presence of starch in­
cromolecules exposed to RS. Additionally, determination of AOX dicator can be applied. This is one of the oldest approaches to
level can also indicate the oxidative status in human body [130]. But measure the amount of total hydroperoxides and oxidative rancidity
increased TAC in plasma or serum sometimes indicates oxidative [133]. There are some modifications reported in the literature on the
stress, for example, elevation of uric acid related to a certain disease determination of liberated I2 (or I3-), such as using potassium iodide-
such as renal failure can elevate TAC [131]. On the other hand, a silica gel reagent [142], conducting potentiometric titration [143]
decrease in the plasma or serum TAC level may cause decreased and automated potentiometric titration. When AOX compounds are
production of RS [132]. In this case, urate-free TAC level of serum present in the reaction medium, the released I2 concentration is
gains importance, as urate is an AOX molecule compensating for a decreased due to the RS scavenging effect of AOXs, thereby leading

15
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

to the indirect determination of AOX activity [144]. Çelik et al. had an AOX efficiency [30]. Bakır et al. used the TBARS test in a linoleic acid
interesting approach in this regard by developing a novel colori­ emulsion model system, and the authors reported that TBARS assay
metric sensor for measuring hydroperoxide content using starch- was not successful to reflect the relationship between the quantity of
stabilized gold nanoparticles (ss-AuNPs); upon the addition of ss- protecting AOXs and the decrease in the amount of formed chro­
AuNPs and excess I- to a hydroperoxide solution, the formed I3- was mophore [157].
rapidly adsorbed on the surface of nanoparticles causing their ag­ To determine lipid peroxidation products, GC-MS is also used. It
gregation with a concomitant red shift (from 525 nm to 563 nm) of is possible to get more accurate results by chromatography com­
surface plasmon resonance (SPR) absorption of AuNPs. The con­ pared to spectrophotometry. However, most of the lipid oxidation
centration of hydroperoxides estimated in linoleic acid peroxidation products are not thermally stable, hence a derivatization should be
correlated well with those found by the reference ferric thiocyanate applied. The other chromatographic techniques used in this field are
assay. The method was useful for measuring both hydroperoxide and electron ionization (EI) MS coupled to selected ion monitoring (SIM)
peroxyl radical scavenging AOXs [145]. technique, electron capture (EC) and atmospheric pressure chemical
Ferric thiocyanate and ferric xylenol orange assays are also used ionization (APCI) MS (EC-APCI-MS) [158,159].
for this purpose. These methods are based on the oxidation of Fe(II)
to Fe(III) in the presence of hydroperoxides. Then, the generated Fe 4.2. Determination of Oxidative Protein Damage
(III) forms colored complexes with thiocyanate [146] and xylenol
orange [147]. The ferric xylenol orange assay is also called the FOX Proteins are the major targets to RS because they contain certain
assay which is used for determination of lipid peroxides in biological amino acid residues and thiol groups vulnerable to oxidation. Due to
samples [148]. As mentioned previously, the color intensity of gen­ the effect of RS on proteins, some modifications occur and these can
erated complexes decreases in the presence of AOX compound in the be used to quantify protein damage. Oxidative damage of proteins
reaction medium [133]. causes some changes in parent amino acid residues which can be
Another analytical method to track lipid peroxidation is ultra­ determined by different analytical techniques such as HPLC,
violet measurement of conjugated dienes. During lipid peroxidation, fluorometry and LC-MS [80]. Another oxidative stress biomarker is
after the formation of lipid radicals, conjugated dienes that exhibit oxidation of low-molecular-mass thiols. For example, GSH can be
π-π * electronic transitions are formed by rearrangement of bonds in oxidized to different products such as GSSG, mixed disulfides or
the molecule. This allows the degree of lipid peroxidation to be glutathione sulfonamide. Glutathione reductases reduce disulfides
determined by measuring the absorbance produced at 230–235 nm to GSH. This change can be monitored spectrophotometrically by
[133]. Conducted spectrophotometric measurement in UV region using 5,5′-dithiobis (2-nitrobenzoic acid) (DTNB, also known as
makes the method susceptible to interference from organic com­ Ellman’s reagent) at 412 nm. Here, GSH, generated from GSSG by
pounds absorbing in the same wavelength range. Therefore, the glutathione reductase, is reacted with DTNB [160]. Erel and Nese­
method needs a pre-separation such as solvent extraction prior to lioglu designed a DTNB reagent−based automated assay for thiol/
analysis of biological samples [149]. The method was also applied for disulfide homeostasis using sodium borohydride as reducing agent
the determination of AOX activity in some synthetic and natural [161]. Görüşük et al. adopted a different approach for determining
samples [150,151]. both thiols and disulfides with the same 2,2'-azinobis-3-ethylben­
The kinetic analysis of lipid peroxidation can provide useful in­ zothiazoline-6-sulfonic acid (ABTS) reagent, where the slower re­
formation on the absolute or relative rate constants for the lipid acting disulfides required an elevated temperature incubation for
peroxyl radical scavenging ability of AOXs. It is also important for reducing the ABTS•+ radical cation [162]. As residues of reducing
understanding the effect of AOX action and its radical scavenging agents pose additional problems in redox-based assays, Çifteci et al.
capacity [140]. developed an AuNPs–based turn-on fluorometric sensor for quanti­
In regard to electrochemical determination of hydroperoxides, fication of sulfhydryl and disulfide forms of biothiols in relation to
Adhoum and Monser developed a Prussian blue (PB)-modified glassy the measurement of thiol/disulfide homeostasis. In this assay, elec­
carbon sensor electrode on which hydroperoxides are electro­ trostatic adsorption of water-soluble fluorophore Rhodamine–6 G
chemically reduced; the method was applied to the amperometric (Rh–6 G) dye onto citrate–stabilized AuNPs resulted in Förster re­
determination of peroxide value (PV) in edible oil. The sensor elec­ sonance energy transfer (FRET) in the turn–off mode, but due to the
trode responded to all hydroperoxides, but the strongest responses high surface affinities of biothiols (in both thiol and disulfide forms)
were recorded for H2O2 followed by tert-butyl hydroperoxide, cu­ to AuNPs, the fluorescence increment at 552 nm (turn-on mode)
mene hydroperoxide and linoleic acid hydroperoxide [152]. accompanying the release of Rh–6 G dye molecules indicated thiol
content. Due to the greater affinity of AuNPs for thiols, the accom­
4.1.2. Determination of Thiobarbituric Acid Reactive panying release of the fluorescent dye Rh-6 G from the nano­
Substances (TBARS) particles’ surface gave rise to an increase in fluorescence. By this
selective adsorption-desorption process onto-from gold nano­
The thiobarbituric acid reactive substances (TBARS) assay is particles, the assay could measure the total amount of thiols and
probably the most common and most criticised method of de­ disulfides, but when free thiols were blocked by the thiol-alkylating
termining lipid oxidation. One of the final products of lipid oxidation agent N-ethylmaleimide, only disulfides gave the fluorescence re­
is MDA. The method is based on measuring the optical absorbance of sponse, thereby enabling to differentiate disulfides from thiols and
the chromogenic adduct compound consisting of MDA and thio­ determine thiol/disulfide ratio (as the wellness indicator) fluor­
barbituric acid (TBA) at 1:2 stoichiometry (MDA-(TBA)2) at 532 nm ometrically [163].
spectrophotometrically [153] or at 553 nm fluorometrically [154]. Except determination of changes on amino acid residues; protein
The basic disadvantage of TBARS method is that, especially in carbonyls, disulfides (−S-S−), sulfinic (−SO2H) or sulfonic (−SO3H)
complex matrices, TBA can react with compounds which are not acids are among the mostly used biomarkers of oxidative protein
directly related to lipid oxidation causing erroneous results damage [140]. Quantification of protein modification by oxidants by
[155,156]. To separate MDA from possible interferents, the method making use of the reactions such as the loss of the parent amino acid
can be combined with FTIR and HPLC to enhance selectivity and residue, formation of unstable intermediates, and the generation of
sensitivity [156]. Similar to the other oxidative stress parameters, the stable products was excellently reviewed by Hawkins et al.; the most
production of MDA decreases in the presence of AOX compounds. common methods to measure protein oxidation products are HPLC-
Therefore, TBARS method can also be used for the determination of MS, GC-MS and Western blot/ELISA test [80].

16
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

The most studied oxidation product is protein carbonyls, which protein, BSA, was oxidized by means of a classical Fenton system
can be analyzed in different biological samples. Their chemical (FeSO4/H2O2). The tested AOXs were four different flavonoids
stability at appropriate storage conditions is another advantage namely; catechin, kaempferol, apigenin and naringenin. All tested
(they are stable for 3 months at −80oC) [164]. One of the well-known polyphenols caused a change in the SWV signal of BSA in the solu­
methods for the determination of protein carbonyls is based tion containing cobalt(III) tris-2,2’-bipyridine complex. The re­
on the reaction between carbonyl compounds produced by protein searchers determined that the most effective AOX among the tested
oxidation and 2,4-dinitrophenylhydrazine (DNPH). The formed compounds was catechin [171].
2,4-dinitrophenylhydrazones can be determined either spectro­
photometrically (λmax = 370 nm) or using Western blotting ELISA 4.3. Determination of Oxidative DNA Damage
techniques [80,165]. However, since the colors of the product
(hydrazone) and the reagent (hydrazine) are nearly identical, and Considering the determination of oxidative DNA damage, the first
other possible interferents (e.g., heme-containing proteins and compound that comes into mind is 8-hydroxy-2-deoxyguanosine (8-
retinoids) absorb light around this analytical wavelength, the OHdG) as the most examined oxidation product, however, it is not
analysis may overestimate the result [166]. Bayarsaikhan et al. the only biomarker of oxidative DNA damage. Oxidative attacks to
developed an alternative modified CUPRAC method to identify DNA can cause simultaneous base damages and modifications.
the products derived from oxidative protein damage and measure Chromatographic methods, especially GC-MS, is often used to detect
the protective effects of different AOXs on this damage. The authors these products [172]. Among the other oxidation damage products of
used three different model proteins, namely bovine serum albumin, DNA bases, 2-hydroxyadenine (2-OH-Ade), 5-OHC, 5-FoU, and 5-
egg white and fetal bovine serum, for this purpose. In this study, OHU can be mentioned [173,174]. In a recent study co-authored by
CUPRAC method was used in two different ways (i.e. in solution and Halliwell and Dizdaroğlu [175] as the two leading researchers in this
on a Nafion membrane sensor). They also used the modified DNPH field, it has been reported that the hydroxyl radical has an important
carbonyl determination method to compare their findings. contribution to oxidative DNA damage in vivo. The authors also ex­
According to this modified DNPH assay, the final anionic product was pressed that hydroxyl radical, unlike all other RS, generates a large
obtained in strongly alkaline medium in the presence of a cationic number of different damage products from all DNA bases. Some of
surfactant (cetyl trimethyl ammonium bromide) and after extraction the other RS either do not react directly with DNA or selectively
into acetonitrile, the absorbance was read at 430 nm [167]. The target guanine due to its relatively low standard reduction potential
presence of concentrated NaOH shifted λmax to longer wavelengths (E0) value. Since DNA has negatively charged phosphate groups,
and allowed a direct quantification without a need for lengthy transition metal cations such as divalent Fe and Cu are bonded to
pretreatments [168]. DNA. On the other hand, H2O2 can cross intracellular membranes
In another technique used by Ma et al., carbonylated proteins and when it reaches DNA, •OH can be generated by Fenton reaction
were determined by frequency-shift based surface-enhanced Raman and causes ‘site-specific’ damage on DNA. The distribution of DNA
spectroscopy (SERS) immunoassay. The authors reported the elim­ degradation products caused by site-specific and other modes of
ination of possible interferences by using a capture antibody [169]. hydroxyl radical attack upon DNA through transition metal io­
There are a number of fluorophore compounds produced at the n−induced •OH generation depends on several factors, including the
end of several oxidative and glycoxidative modifications of amino site of metal binding to DNA [175].
acid residues, giving way to fluorescence spectroscopy to determine During the oxidation of DNA, not only damaged bases but sugar
protein oxidation. The drawback of this method is not to allow in­ modification products are also generated and these can be mon­
dividual analysis of tested compounds as it measures all fluorophore itored by GC-MS or LC-MS techniques. Among other DNA oxidation
products in the reaction medium, meaning that substances that biomarkers are oxidized purines (and pyrimidines), strand breaks,
fluoresce but are not a product of protein oxidation will cause in­ and C-8 hydroxylation products of guanine, 8-oxo-7,8-dihydro-2’-
terference. Thus, in order to separate protein oxidation biomarkers, a deoxyguanosine. Since the amount and types of the oxidation pro­
preliminary chromatographic separation is needed. ducts are closely related to reaction conditions and redox status of
Finally, the overall structure of an oxidized protein can be the cell, assesment of multiple products could give more realistic
monitored by using different approaches such as gel electrophoresis, results [133].
light scattering analysis and X-ray crystallography. Using these Among the most widely used techniques to identify biomarkers
techniques, it is possible to compare intact and oxidized protein associated with DNA oxidation are electrochemical detection
samples, but these approaches are not capable of showing site- methods, namely cyclic voltammetry (CV), differential pulse vol­
specific oxidative damage. tammetry (DPV), and square wave voltammetry (SWV), using bare or
There are also different electroanalytical methods in literature suitably modified electrodes. Especially nanostructured materials
aiming to measure oxidative protein damage. For example, Enache (either carbon− or metal−based) are often used as electrode surface
et al. developed an electrochemical sensor to detect the carbonyl modifiers. There are also studies in which electrodes are directly
groups generated by oxidation of proteins with RS. The method was modified by single-stranded DNA; alternatively, oxidative DNA da­
based on the redox characteristics of DNPH. The model protein used mage can be followed with the use of dendrimer, aptamer or anti­
for the study was bovine serum albumin (BSA). The researchers in­ body modified electrodes to achieve high sensitivity and selectivity
vestigated electrochemical behavior of intact and oxidized BSA using at relatively low cost. Recently, they have been miniaturized and
differential pulse voltammetry technique and glassy carbon elec­ used in automation systems [176].
trode. The authors showed that the hydrazine moiety of the DNPH Some degree of oxidative damage to DNA is often associated with
reagent was the electroactive center and was responsible for car­ age-related cancer. So, oxidative DNA damage can be a meaningful
bonyl complexation. DNPH was immobilized on a mixed 4-styr­ indicator showing a person’s cancer risk. On the other hand, there
enesulfonic acid-Nafion matrix to retain its properties [170]. are ongoing discussions on the possibility of cancer prevention
Sun et al. investigated both protein damage and protective effects through DNA protection against oxidative damage with the aid of
of certain AOX compounds. The authors used the BSA/poly (dia­ AOX compounds taken up via appropriate diets [172].
llyldimethylammonium chloride) (PDDA) functionalized graphene In a sample study, the oxidative damage on DNA and protective
nanosheets (PDDA-G) composite film modified glassy carbon elec­ effects of AOX compounds were determined by using gas chroma­
trode (BSA/PDDA-G/GCE) for this purpose. The electrochemical tography–tandem mass spectrometry. The authors measured a series
method chosen was square wave voltammetry (SWV). The model of DNA oxidation products in the presence and absence of V.vinifera

17
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

Table 1
Biomarkers of oxidative stress and antioxidant defense system in various diseases.

Disease Oxidative stress biomarkers Antioxidant status Reference

Cancer
Breast cancer MDA, AOPP, Trx SOD [181]
Breast cancer TBARS, LH, carbonylation of serum proteins FRAP, GSH [182]
Prostate cancer MDA, NO, TPP, OSI TAC [183]
Prostate cancer TBARS, carbonylation of serum proteins CAT, SOD, vitamin C and E, NPSH [184]
Prostate cancer LPO, 8-OHdG CAT, SOD, GST [185]
Colorectal cancer MDA, ox-LDL TAC, SOD, GPx, GST [186]
Colorectal cancer NOX, XO, AGE, AOPP, MDA, 8-OHdG SOD, CAT, GPx, GR, GSH [187]
Cervical cancer 8-OHdG, carbonylation of serum proteins, MDA SOD, CAT, GPx [188]
Lymphoblastic leukemia TBARS, carbonylation of serum proteins CAT, SOD [189]
Lung cancer TBARS CAT, SOD, TS-H, NPSH
Neurodegenerative disease
Amyotrophic lateral sclerosis MDA, 8-OHdG GPx, GR, SOD, TAS, GSSG, GSH [190]
Parkinson’s disease LPO, MDA, CRP SOD, CAT, TRAP, SH [191]
Parkinson’s disease MDA, AOPP, 4-HNE [192]
Parkinson’s disease AOPP, TBARS SOD, CAT, FRAP, vitamin C, NPSH [193]
Alzheimer's disease TBARS, MDA, homocysteine FRAP [194]
Autism MDA GSH, CAT, PON1, TAC [195]
Diabetes
Type 1 diabetes mellitus MDA, NO, FBS, HbA1c SOD, CAT, GPx [196]
Type 1 diabetes mellitus LPO, MDA, carbonylation of serum proteins, HbA1c GSH, GPx, α-tocopherol, β-carotene [197]
Type 1 diabetes mellitus HbA1c Cu/Zn-SOD, CAT, TAS [198]
Type 2 diabetes mellitus MDA GSH, GR, GPx, SOD, CAT [199]
Type 2 diabetes mellitus LPO, MDA FRAP, protein thiols, GSH, CAT [200]
Gestational diabetes mellitus TOS, OSI Irisin, TAS [201]
Gestational diabetes mellitus MDA, 8IsoP, XO, LPO SOD, TAC, GPx, vitamin C, vitamin E [202]

(var. alphonse) extract rich in AOXs, and concluded that the use of comparing that of control groups. Biomarkers of oxidative stress and
the extract caused a significant decrease in the amounts of DNA AOX defense in various diseases are covered in Table 1.
oxidation products [177]. MDA, malondialdehyde level; SOD, superoxide dismutase ac­
Instead of detecting biomarkers, studies on the determination of tivity; AOPP, advanced oxidation protein product; Trx, thioredoxin;
RS directly by using electrochemical sensors to monitor oxidative GSH, reduced glutathione; TBARS, thiobarbituric acid reactive sub­
stress have been mentioned by Deshpande et al. in a recent review. stances; LH, lipid hydroperoxide; FRAP, ferric reducing antioxidant
Although using glassy carbon electrodes is possible for this purpose, potential; 8-OHdG, 8-hydroxydeoxyguanosine; LPO, lipid hydroper­
there are a number of restrictions for live tissues. The best solutions oxides; GST, glutathione S-transferase; GPx, glutathione peroxidase;
to overcome this situation is to use small sized electrodes (size: ox-LDL, oxidized-low density lipoprotein cholesterol; GR, glu­
5–10 nm, diameter: ~100 mm) having the ability of measuring RS in tathione reductase; AGE, advanced glycation end products; TAC, total
the proximity of cells [41]. For example, using this approach H2O2 antioxidant capacity; TAS, total antioxidant status; NPSH, non-pro­
and O2•– could be detected by means of Au-nanocones modified tein thiols; TS-H, total thiols; GSSG, glutathione disulfide; SH, sulf­
carbon fiber microelectrodes (CFME) in a single drop of blood. Here hydryl group; TRAP, total radical-trapping antioxidant parameter; 4-
the authors used coupling of the small sized CFME and a selective HNE, 4-hydroxynonenal; XO, xanthine oxidase; NOX, NADPH oxi­
synthetic receptor. The technique used here was DPV [178]. dase; NO, nitric oxide; PON1, paraoxonase 1 activity; FBS, fasting
In another example study to determine O2•–, the suitable redox blood sugar; HbA1c, glycated hemoglobin; TOS, total oxidative
potential of O2/O2•– couple vs. NHE (330–140 mV) was exploited. stress; OSI, oxidative stress index; 8IsoP, 8-isoprostane.
This value allowed direct determination of O2•– by electrochemical
oxidation using platinized microelectrodes or with nitrogen-doped 6. Biomarkers of Oxidative Stress and Antioxidant
carbon AgNPs electrodes [179]. Defense in Cancer
Beissenhirtz et al. developed a new type of superoxide sensor
based on a multilayer cytochrome c (cyt. c) electrode. To develop the 6.1. Breast cancer
gold wire electrode, it was modified by alternating layers of cyt. c
and poly(aniline(sulfonic acid)). The authors concluded that the in­ Breast cancer is a major public health issue and is the primary
crease in the layers contributed to electrochemical activation of cyt. reason of cancer related death among women worldwide. In a re­
c. The developed electrode was used in the determination of en­ presentative study on breast cancer, it was stated that oxidative stress
zymatically generated superoxide. The authors concluded that the is responsible from promotion and progression of various cancers,
multilayer sensor showed a superior sensitivity for O2•– determi­ such as breast cancer [203]. Therefore, in order to reveal the asso­
nation compared to monolayer electrode systems. Additionally, the ciation between the disease and oxidative stress biomarker levels, 23
researchers reported the good thermal stability of the modified surgically treated women with breast cancer (against 13 healthy
electrode [180]. controls) were involved in the study [181]. Blood samples of the pa­
tients were analyzed in terms of malondialdehyde (MDA) levels as an
index of lipid peroxidation, superoxide dismutase (SOD) activities,
5. Balance Between Oxidants and Antioxidants advanced oxidation protein product (AOPP) and thioredoxin (Trx)
levels. The results of this study indicated that MDA, SOD, AOPP as well
The aim of this part of the study is to give detailed information Trx levels of the patients were found to be significantly higher than
about oxidative stress and antioxidant status (AS) biomarkers to be those of the controls. As the data pairs of serum parameters in control
measured in each pathogenesis. Then, the balance between oxidative group and breast cancer patients, Trx, MDA, AOPP and SOD levels
stress and AOX defense system in patients were also discussed by were (4.12; 141 ng/mL), (3.08; 6.58 nmol/mL), (60.2; 148.7 µmol/L),

18
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

(6.76; 22.2 Units (U) /mL), respectively, patients having higher levels peroxidase (GPx) and GST levels in one hundred sixty eight patients
than controls in all parameters [181]. Additionally, the oxidative stress with colorectal cancer were investigated. A glance at the results
caused by different treatments during breast cancer was also in­ revealed the significantly higher homocysteine, cysteine, folate and
vestigated in a recent study; the degree of oxidative stress produced glutathione levels in patients with colon or rectal cancer when
by adjuvant chemotherapy, radiotherapy or hormonal therapy treat­ compared to those of control. While higher homocysteine con­
ments in seventy women with breast cancer was examined. After centration was associated with the risk of colorectal cancer; cy­
collecting blood samples, GSH, TBARS, lipid hydroperoxide (LH) level steine, PLP and folate had no effect on the risk of colorectal cancer.
and carbonyl level measurements were conducted as oxidative stress Moreover, 151 colorectal cancer patients had significantly higher ox-
analysis, and FRAP assay was used to detect serum TAC levels. The LDL levels and SOD activities as well as significantly lower GPx and
FRAP values were associated with diet quality only at the baseline (at GST activities than those of 188 healthy controls. In the AOX defense
p0); oxidative stress parameters showed an increase after adjuvant system, SOD and GPx are considered to be the primary AOX enzymes
chemotherapy (at p1), but they were not associated with diet and GST plays as the secondary AOX enzymes. The raised SOD ac­
quality [182]. tivity could catalyze the dismutation of superoxide into oxygen and
hydrogen peroxide. Later, catalase and GPx activities manage the
6.2. Prostate cancer elimination of hydrogen peroxide. However, in this study, excessive
amount of hydrogen peroxide or cellular toxicity of cancer resulted
Prostate cancer is another serious worldwide health problem and with the decreased GPx and GST activities [186]. These results are in
it is the second most common malignancy in aging men [204]. Due agreement with Zińczuk et al. [187] who showed that there is an
to the fact that the cause of oxidative stress relies on an imbalance association between the subjects with colorectal cancer and enzy­
between reactive oxygen/nitrogen species or lipid peroxidation matic/non-enzymatic redox imbalance, increased oxidative damage
products and the cell’s detoxification defense system [205], a case- to proteins, lipids and DNA. Redox homeostasis was defined as the
control study was designed with 120 patients with prostate cancer balance between TAC and total oxidant status (TOS). For the ex­
(against 100 healthy controls) to assess the levels of blood MDA, NO, amination of redox status, pro-oxidant enzymes including NADPH
total plasma peroxide (TPP), TAC as well as oxidative stress index oxidase (NOX) and XO; AOX enzymes such as SOD, CAT, GPx, glu­
(OSI) [183]. Results showed with statistical significance (set at tathione reductase (GR) and GSH as well as oxidative damage pro­
p < 0.05) that the patients with prostate cancer had significantly ducts like advanced glycation end products (AGE), AOPP, MDA and 8-
higher MDA (0.48 and 0.29 µmol/L respectively), NO (4.6 and OHdG were determined in normal and cancerous tissue of twenty-
2.4 µmol/L, respectively), TPP (25.3 and 17.2 µmol/L H2O2, respec­ nine patients with colorectal cancer. According to the results, NOX,
tively) and lower TAC levels (3.5 and 3.9 mmol/L, respectively) in XO, SOD, CAT and TAC levels as well as oxidative damage products
comparison with control groups. In order to investigate the presence were found to be significantly higher in cancer tissue than in normal
or absence of bone metastasis in the levels of oxidative damage and colon mucosa [187].
AOX defense, biological damage caused by free radicals in lipids and
proteins was determined by TBARS and carbonylation of serum 6.4. Cervical cancer
proteins in 55 patients with prostate cancer against 55 healthy men.
Moreover, AS of the patients was examined by enzymatic (CAT and With regard to serum oxidative stress and AOX biomarker levels
SOD) and non-enzymatic AOXs (non-protein thiols (NPSH), vitamin in cervical cancer patients, 8-OHdG, carbonylation of serum proteins
C and vitamin E) [184]. Results indicated that TBARS and protein and MDA of oxidative damage products as well as SOD, CAT, GPx, and
carbonyl levels were increased for both patients with localized total AS of AOX defense mechanisms were studied. Results high­
prostate cancer and bone metastatic disease. While SOD activity was lighted the significant increase in oxidative damage markers and
increased only in patients with localized prostate cancer, NPSH le­ significant decrease in AOX parameters in cervical cancer patients
vels were increased in patients with bone metastasis. Besides, vi­ compared to controls [188].
tamin C and E contents as well as CAT activity were reduced in both
groups in comparison to the control. Similar findings were reported 6.5. Leukemia
by Shukla et al. [185], indicating higher 8-OHdG levels to quantify
the DNA damage and lipid hydroperoxides (LH) as well as lower GST Similar trends were obtained for lymphoblastic leukemia pa­
value. However, there was no significant variation in CAT and SOD tients [189]. Increased oxidative damage products including TBARS
levels [185]. and serum protein carbonyls and reduced AOX levels (CAT, SOD)
were found in the patients compared to controls.
6.3. Colorectal cancer On the other hand, Zanini et al. [210] reported higher non-en­
zymatic thiols (NPSH and total thiol groups (TS-H)) levels and an­
Colorectal cancer is the third and the second most frequent type tioxidant enzymes (CAT and SOD) activities as well as lower TBARS
of cancer in men and women, respectively [206]. It has been stated values in the patients compared to controls. It was explained by
that increased homocysteine concentration [207], higher levels of increased tumor size, progression of cancer and overproduction of
oxidative stress and increased or decreased AOX enzyme activities ROS, because it can be assumed that rapidly dividing cells are prone
are among the risk factors of colorectal cancer [208]. It has been to build an oxidant-antioxidant status appropriate for their growth;
indicated that hyperhomocysteinemia (homocysteine concentration for instance, high level of AOX and low substrate of peroxid­
≥ 14 µmol/L) admits overproduction of free radicals and hydrogen ability [211].
peroxide, and may further cause gene mutation and cancer. In ad­
dition to this, high homocysteine level also affects cysteine which is 7. Biomarkers of Oxidative Stress and Antioxidant Defense in
responsible for the rate-limiting impact on the synthesis of GSH and Neurodegenerative Diseases
GSH-dependent AOX defense system [209]. In a study on the re­
lationship between high homocysteine levels and increased risk of 7.1. Amyotrophic lateral sclerosis (ALS)
colorectal cancer [186], plasma homocysteine, cysteine, pyridoxal 5’-
phosphate (PLP), folate and reduced glutathione values, oxidative Amyotrophic lateral sclerosis (ALS) is the most common motor
stress indicators including MDA and oxidized-low density lipopro­ neuron disease and defined by degeneration of upper and lower
tein cholesterol as well as AS measuring TAC, SOD, glutathione motor neurons in the brain and spinal cord [212]. Blasco et al. [190]

19
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

carried out a pilot study with ten ALS patients to present a link there is an increasing risk in the formation of dementia according to
between ALS disease and oxidative stress by measuring GPx, GR, the transition from mild stage to moderate and severe stages of
SOD, TAS, MDA, 8-OHdG, glutathione status (e.g. glutathione dis­ cognitive disruption [218]. In this context, Negahdar et al. [194]
ulfide (GSSG)/reduced glutathione (GSH)), vitamins (B9, B12, A, E, C) analyzed the risk factors in one hundred twenty patients with mild
and metals (Cu, Zn, Mn, Se). The authors reported a significant in­ or moderate to severe cognitive impairment. Serum oxidant/anti­
crease in 8-OHdG and MDA levels and a significant decrease in TAS oxidant status were determined by measuring trace elements
levels of ALS patients compared to controls, indicating an imbalance (copper, manganese and zinc), homocysteine, TBARS, MDA and FRAP
between the production of ROS and the defense system to remove or levels. Results showed that there is an association between the in­
repair the damage. On the other hand, there was no significant creased oxidative stress as well as decreased AS and cognitive im­
change in AOX enzyme activities (SOD, GPx, GR), vitamin and mi­ pairment stage. In addition to these, there was an increasing trend in
neral values, however, glutathione status was altered in ALS patients homocysteine levels, which is responsible for oxidative stress, de­
resulting in a significantly higher GSSG/GSH ratio. Reduced GSH level pending on aging. However, there was no statistical variation in the
is related with an increase in its oxidized form, which could ad­ levels of trace elements in both patients and control group [194].
versely affect the disease evolution [213]. The results of this study
showed that oxidative stress is an appropriate factor in the evalua­ 7.4. Autism spectrum disorder
tion of ALS [190].
In addition to ALS, PD and AD, autism spectrum disorder (ASD) is
7.2. Parkinson’s disease (PD) another neuro developmental disorder and characterized by diffi­
culties in communication and social interaction, repetitive and ste­
PD is the second most common neurodegenerative disorder, in reotypic behaviors and defects in cognitive abilities [219]. It was
which the lowered dopamine levels as well as motor and non-motor reported that oxidative stress, genetic and environmental factors as
manifestations occur due to the injured dopaminergic neurons in the well as immune dysregulation have a role in this disorder [220].
substantia nigra pars compacta [214]. In addition to the oxidation of Abdel-Salam et al. [195] indicated a significant increase in MDA le­
dopamine by monoamine oxidase, chronic inflammatory process vels and decrease in AS, including CAT, paraoxonase 1 (PON1), GSH
and high concentrations of iron in the brain lead to RS generation and TAC values in the serum of 3 − 12 years aged individuals with
and cellular dysfunction, resulting in neurodegenerative diseases autism spectrum disorder compared to the control group. Moreover,
[215]. Since the increased damage to lipid, protein and DNA might be nuclear factor kappa B (NF-κB), which is a protein transcription
tracers of oxidative/ nitrosative stress in this pathogenesis [192], factor, was found to be significantly higher in children with autism
current studies aim to investigate the relationship between oxida­ rather than that of control group, suggesting activation of NF-κB
tive/nitrosative stress parameters and AOX defense status. For in­ with increased status of oxidative stress [195].
stance; de Farias et al. [191] indicated a higher correlation between
increased LPO, MDA, SOD activities as well as lower total TRAP, 8. Biomarkers of Oxidative Stress and Antioxidant
sulfhydryl group (SH), CAT activity and PD disease. It has been re­ Defense in Diabetes
ported that higher lipid peroxidation indicators, including LPO and
MDA, are linked to lower AOX concentrations, high substrate levels Diabetes mellitus (DM), one of the major disorders, is char­
for oxidation and unidentified plasma pro-oxidants in cases with PD. acterized by hyperglycemia along with carbohydrate, lipid, and
Moreover, while enhanced SOD activity may arise from higher pro­ protein metabolism abnormalities [221]. DM is predicted to affect
duction of superoxide radicals, resulting in scavenging of ROS but 592 million people by 2035 [222]. It was reported that one of the
H2O2 generation, lower levels of CAT might be detected due to in­ pathogenesis associated with diabetes may be related to oxidative
creased usage to decompose H2O2 [191]. With regard to analyze stress [223]. Free radicals that are produced at elevated amounts in
oxidative stress−antioxidant defense balance in PD subjects, the diabetes owing to glucose autoxidation process as well as simulta­
oxidative stress biomarkers such as MDA, AOPP and 4-HNE as well as neous decrease in AS may deteriorate the lipids, proteins, cellular
prooxidant–antioxidant balance were determined. Results indicated organelles and DNA, resulting in insulin resistance [224,225].
that these markers are found to be gender- and age-related [192]
Indeed, it was reported that elder male subjects are more susceptible 8.1. Type 1 diabetes mellitus
to this disease [216]. In another study, Medeiros et al. [193] explored
the peripheral iron, ferritin, and transferrin levels, oxidative stress Type 1 diabetes mellitus (T1DM) is an autoimmune disorder
biomarkers and AOX activity in PD patients. Results revealed that caused by defect of pancreatic–beta cells [226] and involves ap­
iron levels were found to be significantly lower in PD individuals proximately 5–10% of all diabetes cases [196]. In a recent study, as­
(67.5 μg/dL) rather than that of controls (78 μg/dL), whereas there sociation between oxidative stress levels including MDA, NO, fasting
was no significant difference in ferritin and transferrin concentra­ blood sugar (FBS) and glycated hemoglobin (HbA1c) and AOX en­
tions. Although high level of iron in the substantia nigra is regarded zyme status such as SOD, CAT and GPx in T1DM patients were
as a risk factor in PD, decreased level in peripheral blood is possibly a analyzed. Results indicated that significantly higher FBS
problem for the function of neuronal enzymes as well as ferritin (173.09 ± 17.80 mg/dL) and HbA1c concentrations as well as MDA
reservoir in neurons, causing accumulation of free iron in the sub­ and NO levels were obtained in T1DM cases rather than those of
stantia nigra [217]. Furthermore, decreased AS including vitamin C, healthy individuals. Moreover, serum CAT, SOD and GPx activities
NPSH and FRAP and increased oxidative stress levels such as TBARS were found to be statistically lower by 16.7%, 72.8%, and 15.3%, re­
and AOPP were obtained in the PD cases in comparison with con­ spectively in T1DM subjects in comparison with those in controls,
trols. On the other hand, there were no significant variation in SOD indicating a decrease in the AOX enzyme activities due to glycation
and CAT activities of PD patients and controls [193]. of enzymatic proteins [196]. Similarly, Codoñer-Franch et al. [197]
investigated oxidative stress by measuring the primary (LPO) and
7.3. Alzheimer’s disease (AD) secondary (MDA) lipid peroxidation products as well as level of
carbonylation of serum proteins. While endogenous AOX defense
AD is an irreversible brain disorder, which could be induced by system was evaluated by determining the GSH and GPx, exogenous
dyshomeostasis of biochemical parameters. AD is the most common AS was determined by means of α-tocopherol and β-carotene. Sta­
form of irreversible dementia pertaining to elderly stages of life, and tistically elevated levels of lipid (LPO and MDA) and protein

20
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

(carbonylation of serum proteins) oxidation markers were detected levels, insulin sensitivity and low-grade inflammation, were found to
in the patients with T1DM in comparison with control groups. Be­ be significantly lower in the GDM patients in comparison with
sides, GPx activity, which is responsible from protection of cell control group [201]. Besides, Shang et al. [202] investigated the AOX
proteins and membranes against oxidative damage, were decreased defense system and oxidative stress in pregnant woment with GDM
in T1DM patients, whereas there was no variation in the GSH con­ by measuring MDA, 8-isoprostane (8IsoP), XO, SOD and TAC levels in
centration among the groups. With respect to the results of exo­ maternal, cord and placenta. Results demonstrated that oxidative
genous AOX activities, lower α-tocopherol/total Ch ratio and β- stress status including MDA, XO and 8IsoP amounts were found to be
carotene concentrations were found in the subjects with T1DM significantly increased, while AOX defense system in terms of SOD
when compared with control groups, suggesting usage of AOXs due and TAC levels were significantly decreased in subjects with GDM
to higher oxidative stress [197]. In another study, Salmonowicz et al. compared to the control group [202].
[198] investigated the effects of trace elements which have an im­
pact on the AOX defense system in children with T1DM. According to 9. Conclusions and Perspectives
the results, participants with T1DM showed lower levels of Mg
(19.27 μg/mL), Zn (88.35 μg/100 mL), higher concentrations of Cu Because of their instability, RS are often measured by indirect
(126.19 μg/100 mL) and decreased TAS (0.894 mmol/L) levels com­ methods. An excess of RS, which cannot be counter-balanced by AOX
pared to those of of control groups (20.54 μg/mL Mg, 97.00 μg/ compounds, causes damage to biomacromolecules such as DNA,
100 mL Zn, 110.55 μg/100 mL Cu and 1.170 mmol/L TAS), suggesting a proteins and lipids in the body. One of the most common strategies
relation between damaged AOX defense system and deficiency of Mg for detection and quantification of RS is to measure their damaging
and Zn and increased Cu levels (the indicated metal ions may be effects on these biomacromolecules called oxidative stress (OS)
assumed to be divalent, though not indicated in the source litera­ biomarkers, where antioxidant defenses can help protect these
ture) [198]. biomarkers from oxidative damage. There is a clear (inverse) re­
lationship between increasing amount of RS and decreasing AOX
8.2. Type 2 diabetes mellitus concentration. OS has been shown to be associated with the pro­
gression of various diseases including diabetes, neurodegenerative
Recently, it has been reported that complications associated with disorders and cancer, and OS is best reflected in the observed oxi­
generation of T2DM may be related to the same oxidative stress dative modifications on biomacromolecules, opening the way to
pathways [227]. In the context of the relation between AS and oxi­ better diagnose these diseases before they are clinically evident. The
dative stress levels in patients with T2DM, Siddiqui et al. [199] de­ clinical use of in vitro tests on biomarker concentrations and TAC in
monstrated that participants with newly diagnosed T2DM had direct disease diagnosis is a controversial issue. However, it is gen­
significantly higher amount of MDA (2.66 ± 1.11 nmol/mL), in­ erally an established fact that oxidative stress parameters rise and
creased GR (1.78 ± 0.85 U/mL) and SOD activity (867.65 ± 259.08 AOX defense parameters decline in certain OS−originated diseases. A
units per gram of haemoglobin, U/gHb) and lower GSH levels particular concentration level of biomarkers can provide useful in­
(15.53 ± 3.78 μmol/gHb) than those of the subjects (2.13 ± 0.72 formation about a person’s health status. Although it is difficult to
nmol/mL MDA, 1.57 ± 0.78 U/mL GR, 754.11 ± 170.75 U/gHb SOD make generalizations on large groups, the sudden change in the
and 17.35 ± 3.6 μmol/gHb GSH) with normal glucose tolerance [199]. concentrations of these substances, especially for a given individual,
Glycated plasma proteins possess a crucial role in diabetes [228]. may provide important clues for diagnosis and treatment of OS-
This process causes formation of different glycation intermediate originated diseases. On the other hand, both RS and TAC are im­
products and protein modifications depending on the glycation portant issues for a variety of scientific disciplines, including bio­
stage, including fructosamine, amino or carbonyl intermediates, chemists, clinical chemists and food chemists. Although several
advanced glycation end-products and amyloid fibrils [228–230]. techniques like UV–vis spectrometry, fluorometry, HPLC and GC with
Tupe et al. [200] investigated the glycated plasma proteins from MS, electrochemical or diode array detectors, ESR/EPR, etc. have been
various glycation stages, oxidative stress and AOX defense system used to detect the specific biomarkers of the oxidative stress, there is
parameters in the patients with T2DM. Results showed that while no single instrumental method to meet all requirements. The same is
there was no significant variation in fructosamine levels, protein true for the selection of biomarkers for specific purposes. In prin­
carbonyl content, AGEs and plasma amyloid groups were sig­ ciple, OS-biomarkers seem to concern mostly food chemistry and
nificantly increased in the patient group compared to the control biochemistry, but this concept can be used for almost all materials
group. Moreover, AS detected as protein thiols and FRAP were found prone to oxidative damage. Although kept beyond the scope of this
to be lower in the diabetic group [200]. review essentially focused on in vitro tests measuring oxidative
stress and related antioxidant defense, in vivo OS-biomarkers may be
8.3. Gestational diabetes mellitus envisaged to gain more popularity in the future.

Gestational diabetes mellitus (GDM) has an increasing pre­ Funding


valence in pregnant women, which is characterized by abnormal
blood sugar levels, leading to fetal abnormalities and maternal This research did not receive any specific grant from funding
complications [231]. In addition to these, GDM may induce gesta­ agencies in the public, commercial, or not-for-profit sectors.
tional hypertension, preeclampsia, fetal macrosomia, shoulder dys­
tocia and rate of cesarean [231,232]. Although the pathophysiology
Declaration of Competing Interest
of GDM has not been clearly understood, it has been indicated that
there is a link between oxidative stress status and GDM-related in­
The authors declare that they have no known competing fi­
sulin resistance among pregnant women, as in T1DM and T2DM
nancial interests or personal relationships that could have appeared
[233]. Therefore, there has been various studies that examine the
to influence the work reported in this paper.
association between oxidative stress and AS in pregnant women
with GDM. For instance; Usluoğullari et al. [201] showed that par­
References
ticipants with GDM had significantly higher serum OGTT (oral glu­
cose tolerance test), OSI, TOS and insulin levels, whereas TAS and [1] B. Halliwell, Free radicals and antioxidants - Quo vadis? Trends Pharmacol. Sci.
irisin content, which has a role in the regulation of glucose and lipid 32 (2011) 125–130 https://doi.org/10.1016/j.tips.2010.12.002.

21
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

[2] İ. Gulcin, Antioxidants and antioxidant methods: an updated overview, Arch. [33] J.M.C. Gutteridge, B. Halliwell, Antioxidants: Molecules, medicines, and myths,
Toxicol. 94 (2020) 651–715, https://doi.org/10.1007/S00204-020-02689-3 Biochem. Biophys. Res. Commun. 393 (2010) 561–564, https://doi.org/10.1016/j.
[3] B. Halliwell, J.M.C. Gutteridge, Free Radicals in Biology and Medicine, 4th ed., bbrc.2010.02.071
Oxford University Press, Oxford, 2007 https://doi.org/10.1093/acprof:oso/ [34] O.I. Aruoma, B. Halliwell, G. Williamson, In vitro methods for characterizing
9780198717478.001.0001. potential prooxidant and antioxidant actions of nonnutritive substances in
[4] J.G. Paithankar, S. Saini, S. Dwivedi, A. Sharma, D.K. Chowdhuri, Heavy metal plant foods, in: O.I. Aruoma, S.I. Cuppett (Eds.), In Antioxidant Methodology,
associated health hazards: An interplay of oxidative stress and signal trans­ AOCS Press, Champaign, Illinois, 1997, pp. 173–204.
duction, Chemosphere 262 (2021) 128350, https://doi.org/10.1016/j. [35] S.D. Çekiç, A. Çetinkaya, A.N. Avan, R. Apak, Correlation of total antioxidant
chemosphere.2020.128350 capacity with reactive oxygen species (ROS) consumption measured by oxi­
[5] S. Ore, Oxidative stress relaxation of natural rubber vulcanized with di-tertiary- dative conversion, J. Agric. Food Chem. 61 (2013) 5260–5270, https://doi.org/
butyl peroxide, Rubber Chem. Technol. 29 (1956) 1043–1046, https://doi.org/ 10.1021/jf3051297
10.5254/1.3542575 [36] H. Bean, F. Radu, E. De, C. Schuler, R.E. Leggett, R.M. Levin, Comparative eva­
[6] N.V. Paniker, S.K. Srivastava, E. Beutler, Glutathione metabolism of the red cells luation of antioxidant reactivity within obstructed and control rabbit urinary
effect of glutathione reductase deficiency on the stimulation of hexose bladder tissue using FRAP and CUPRAC assays, Mol. Cell. Biochem. 323 (2009)
monophosphate shunt under oxidative stress, BBA - Gen. Subj. 215 (1970) 139–142, https://doi.org/10.1007/s11010-008-9972-5
456–460, https://doi.org/10.1016/0304-4165(70)90096-6 [37] R. Apak, K. Güçlü, M. Özyürek, S.E. Karademir, Novel total antioxidant ca­
[7] H. Sies, E. Cadenas, Oxidative stress: damage to intact cells and organs, Philos. pacity index for dietary polyphenols and vitamins C and E, using their
Trans. R. Soc. Lond. B. Biol. Sci. 311 (1985) 617–631, https://doi.org/10.1098/rstb. cupric ion reducing capability in the presence of neocuproine: CUPRAC
1985.0168 method, J. Agric. Food Chem. 52 (2004) 7970–7981, https://doi.org/10.
[8] H. Sies, Oxidative stress: Introductory remarks, in: H. Sies (Ed.), Oxidative 1021/jf048741x
Stress, Academic Press, 1985, pp. 1–8, https://doi.org/10.1016/b978-0-12- [38] I.F.F. Benzie, J.J. Strain, The ferric reducing ability of plasma (FRAP) as a measure
642760-8.50005-3 of “antioxidant power”: The FRAP assay, Anal. Biochem. 239 (1996) 70–76,
[9] H. Sies, Oxidative stress: Concept and some practical aspects, Antioxidants 9 https://doi.org/10.1006/abio.1996.0292
(2020) 852, https://doi.org/10.3390/ANTIOX9090852 [39] J.W. Finley, A.-N. Kong, K.J. Hintze, E.H. Jeffery, L.L. Ji, X.G. Lei, Antioxidants in
[10] C. López-Alarcón, A. Denicola, Evaluating the antioxidant capacity of natural foods: State of the science important to the food industry, J. Agric. Food Chem.
products: A review on chemical and cellular-based assays, Anal. Chim. Acta 763 59 (2011) 6837–6846, https://doi.org/10.1021/JF2013875
(2013) 1–10, https://doi.org/10.1016/j.aca.2012.11.051 [40] S.D. Çekiç, A.N. Avan, S. Uzunboy, R. Apak, A colourimetric sensor for the si­
[11] A.M. Pisoschi, A. Pop, The role of antioxidants in the chemistry of oxidative multaneous determination of oxidative status and antioxidant activity on the
stress: A review, Eur. J. Med. Chem. 97 (2015) 55–74, https://doi.org/10.1016/j. same membrane: N,N-Dimethyl-p-phenylene diamine (DMPD) on Nafion, Anal.
ejmech.2015.04.040 Chim. Acta 865 (2015) 60–70, https://doi.org/10.1016/j.aca.2015.01.041
[12] D. Giustarini, I. Dalle-Donne, D. Tsikas, R. Rossi, Oxidative stress and [41] A.S. Deshpande, W. Muraoka, S. Andreescu, Electrochemical sensors for oxi­
human diseases: Origin, link, measurement, mechanisms, and biomarkers, dative stress monitoring, Curr. Opin. Electrochem 29 (2021) 100809, https://
Crit. Rev. Clin. Lab. Sci. 46 (2009) 241–281, https://doi.org/10.3109/ doi.org/10.1016/J.COELEC.2021.100809
10408360903142326 [42] J. Frijhoff, P.G. Winyard, N. Zarkovic, S.S. Davies, R. Stocker, D. Cheng,
[13] E.A. Rachmilewitz, P.J. Giardina, How I treat thalassemia, Blood 118 (2011) A.R. Knight, E.L. Taylor, J. Oettrich, T. Ruskovska, A.C. Gasparovic, A. Cuadrado,
3479–3488, https://doi.org/10.1182/BLOOD-2010-08-300335 D. Weber, H.E. Poulsen, T. Grune, H.H.H.W. Schmidt, P. Ghezzi, Clinical re­
[14] R. Prabhu, V. Prabhu, R.S. Prabhu, Iron overload in beta Thalasemia-a review, J. levance of biomarkers of oxidative stress, Antioxid. Redox Signal 23 (2015)
Biosci. Tech. 1 (2009) 20–31. 1144–1170, https://doi.org/10.1089/ars.2015.6317
[15] A. Tantiworawit, P. Charoenkwan, S. Hantrakool, W. Choeyprasert, [43] E. Offord, G. van Poppel, R. Tyrrell, Markers of oxidative damage and anti­
C. Sivasomboon, T. Sanguansermsri, Iron overload in non-transfusion-depen­ oxidant protection: current status and relevance to disease, Free Radic. Res 33
dent thalassemia: association with genotype and clinical risk factors, Int. J. (2000) S5–S19.
Hematol. 103 (2016) 643–648, https://doi.org/10.1007/S12185-016-1991-5 [44] M. Anbar, P. Neta, A compilation of specific bimolecular rate constants for the
[16] C. Nathan, A. Cunningham-Bussel, Beyond oxidative stress: An immunologist’s reactions of hydrated electrons, hydrogen atoms and hydroxyl radicals with
guide to reactive oxygen species, Nat. Rev. Immunol. 13 (2013) 349–361, inorganic and organic compounds in aqueous solution, Int. J. Appl. Radiat. Isot.
https://doi.org/10.1038/nri3423 18 (1967) 493–523, https://doi.org/10.1016/0020-708X(67)90115-9
[17] C.C. Winterbourn, Toxicity of iron and hydrogen peroxide: the Fenton reaction, [45] P. Ghezzi, Environmental risk factors and their footprints in vivo – A proposal
Toxicol. Lett. 82–83 (1995) 969–974, https://doi.org/10.1016/0378-4274(95) for the classification of oxidative stress biomarkers, Redox Biol. 34 (2020)
03532-X 101442, https://doi.org/10.1016/j.redox.2020.101442
[18] B. Halliwell, Superoxide-dependent formation of hydroxyl radicals in the pre­ [46] F. Ito, Y. Sono, T. Ito, Measurement and clinical significance of lipid peroxidation
sence of iron chelates, FEBS Lett. 92 (1978) 321–326, https://doi.org/10.1016/ as a biomarker of oxidative stress: Oxidative stress in diabetes, atherosclerosis,
0014-5793(78)80779-0 and chronic inflammation, Antioxidants 8 (2019) 72, https://doi.org/10.3390/
[19] D.T. Sawyer, J.S. Valentine, How super is superoxide? Acc. Chem. Res. 14 (1981) antiox8030072
393–400, https://doi.org/10.1021/AR00072A005 [47] M.S. Shchepinov, V.A. Roginsky, J.T. Brenna, R.J. Molinari, R. To, H. Tsui,
[20] J.P. Klinman, How do enzymes activate oxygen without inactivating them­ C.F. Clarke, A.B. Manning-Boğ, Deuterium protection of polyunsaturated fatty
selves? Acc. Chem. Res. 40 (2007) 325–333, https://doi.org/10.1021/AR6000507 acids against lipid peroxidation, in: R. Watson (Ed.), Omega-3 Fatty Acids in
[21] J.M.C. Gutteridge, Biological origin of free radicals, and mechanisms of anti­ Brain and Neurological Health, Elsevier, 2014, pp. 373–383, https://doi.org/10.
oxidant protection, Chem. Biol. Interact. 91 (1994) 133–140, https://doi.org/10. 1016/b978-0-12-410527-0.00031-4
1016/0009-2797(94)90033-7 [48] D.R. Johnson, E.A. Decker, The role of oxygen in lipid oxidation reactions: A
[22] R. Kohen, A. Nyska, Oxidation of biological systems: Oxidative stress review, Annu. Rev. Food Sci. Technol. 6 (2015) 171–190, https://doi.org/10.1146/
phenomena, antioxidants, redox reactions, and methods for their quantifi­ ANNUREV-FOOD-022814-015532
cation, Toxicol. Pathol. 30 (2002) 620–650, https://doi.org/10.1080/ [49] E. Niki, Do antioxidants impair signaling by reactive oxygen species and lipid
01926230290166724 oxidation products? FEBS Lett. 586 (2012) 3767–3770, https://doi.org/10.1016/j.
[23] W. Dröge, Free radicals in the physiological control of cell function, Physiol. Rev. febslet.2012.09.025
82 (2002) 47–95, https://doi.org/10.1152/physrev.00018.2001 [50] F. Lowe, Biomarkers of oxidative stress, in: I. Laher (Ed.), Systems Biology of
[24] M. Valko, K. Jomova, C.J. Rhodes, K. Kuča, K. Musílek, Redox- and non-redox- Free Radicals and Antioxidants, Springer, Berlin, Heidelberg, 2014, pp. 65–87,
metal-induced formation of free radicals and their role in human disease, Arch. https://doi.org/10.1007/978-3-642-30018-9_4
Toxicol. 90 (2016) 1–37, https://doi.org/10.1007/S00204-015-1579-5 [51] E. Niki, Biomarkers of lipid peroxidation in clinical material, Biochim. Biophys.
[25] B. Halliwell, Free radicals and antioxidants: Updating a personal view, Nutr. Acta - Gen. Subj. 1840 (2014) 809–817, https://doi.org/10.1016/j.bbagen.2013.
Rev. 70 (2012) 257–265, https://doi.org/10.1111/j.1753-4887.2012.00476.x 03.020
[26] M.B. Reddy, L. Clark, Iron, oxidative stress, and disease risk, Nutr. Rev. 62 (2004) [52] H. Yin, L. Xu, N.A. Porter, Free radical lipid peroxidation: Mechanisms and
120–124, https://doi.org/10.1111/J.1753-4887.2004.TB00033.X analysis, Chem. Rev. 111 (2011) 5944–5972, https://doi.org/10.1021/cr200084z
[27] W.H. Koppenol, Generation and thermodynamic properties of oxyradicals, in: [53] C.A. Rice-Evans, Formation of free radicals and mechanisms of action in normal
C. Vigo-Pelfrey (Ed.), Focus on Membrane Lipid Oxidation, CRC Press, Boca biochemical processes and pathological states (Amsterdam), in: C.A. Rice-
Raton, FL, 1989, pp. 1–13. Evans, R.H. Burdon (Eds.), New Comprehensive Biochemistry, Elsevier, 1994,
[28] S. Goldstein, D. Meyerstein, G. Czapski, The Fenton reagents, Free Radic. Biol. pp. 131–153, https://doi.org/10.1016/S0167-7306(08)60441-X (Amsterdam).
Med. 15 (1993) 435–445, https://doi.org/10.1016/0891-5849(93)90043-T [54] M.M. Gaschler, B.R. Stockwell, Lipid peroxidation in cell death, Biochem.
[29] R. Apak, Current issues in antioxidant measurement, J. Agric. Food Chem. 67 Biophys. Res. Commun. 482 (2017) 419–425, https://doi.org/10.1016/j.bbrc.
(2019) 9187–9202, https://doi.org/10.1021/acs.jafc.9b03657 2016.10.086
[30] F. Shahidi, Y. Zhong, Measurement of antioxidant activity, J. Funct. Foods 18 [55] S.N. Desai, F.F. Farris, S.D. Ray, Lipid Peroxidation, Encyclopedia of Toxicology,
(2015) 757–781, https://doi.org/10.1016/j.jff.2015.01.047 Third edition., Elsevier, 2014, pp. 89–93, https://doi.org/10.1016/B978-0-12-
[31] J.M.C. Gutteridge, B. Halliwell, Mini-Review: Oxidative stress, redox stress or 386454-3.00327-4
redox success? Biochem. Biophys. Res. Commun. 502 (2018) 183–186, https:// [56] A. Ayala, M.F. Muñoz, S. Argüelles, Lipid peroxidation: Production, metabolism,
doi.org/10.1016/j.bbrc.2018.05.045 and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal
[32] B. Halliwell, The antioxidant paradox: less paradoxical now?, Br, J. Clin. Pharm. (Article ID), Oxid. Med. Cell. Longev. (2014) (2014) 360438, https://doi.org/10.
75 (2012) 637–644, https://doi.org/10.1111/j.1365-2125.2012.04272.x 1155/2014/360438

22
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

[57] I.F.F. Benzie, Lipid peroxidation: A review of causes, consequences, measure­ [81] W. Zhang, S. Xiao, D.U. Ahn, Protein oxidation: Basic principles and implications
ment and dietary influences, Int. J. Food Sci. Nutr. 47 (1996) 233–261, https:// for meat quality, Crit. Rev. Food Sci. Nutr. 53 (2013) 1191–1201, https://doi.org/
doi.org/10.3109/09637489609012586 10.1080/10408398.2011.577540
[58] M. Shichiri, Y. Yoshida, E. Niki, Unregulated lipid peroxidation in neurological [82] M.J. Davies, Protein oxidation and peroxidation, Biochem. J. 473 (2016)
dysfunction, in: R.R. Watson, F. De Meester (Eds.), Omega-3 Fatty Acids in Brain 805–825, https://doi.org/10.1042/BJ20151227
and Neurological Health, Elsevier, 2014, pp. 31–55, https://doi.org/10.1016/ [83] H.R. Griffiths, Antioxidants and protein oxidation, Free Radic. Res. 33 Suppl
b978-0-12-410527-0.00004-1 (2000) S47–S58.
[59] S. Basu, Review isoprostanes: Novel bioactive products of lipid peroxidation, [84] E.R. Stadtman, B.S. Berlett, Reactive oxygen-mediated protein oxidation in
Free Radic. Res. 38 (2004) 105–122, https://doi.org/10.1080/ aging and disease, Drug Metab. Rev. 30 (1998) 225–243, https://doi.org/10.
10715760310001646895 3109/03602539808996310
[60] M. Janicka, A. Kot-Wasik, J. Kot, J. Namieśnik, Isoprostanes-biomarkers of lipid [85] M.Y. Aksenov, M.V. Aksenova, D.A. Butterfield, J.W. Geddes, W.R. Markesbery,
peroxidation: Their utility in evaluating oxidative stress and analysis, Int. J. Mol. Protein oxidation in the brain in Alzheimer’s disease, Neuroscience 103 (2001)
Sci. 11 (2010) 4659, https://doi.org/10.3390/IJMS11114631 373–383, https://doi.org/10.1016/S0306-4522(00)00580-7
[61] Á. Sánchez-Illana, S. Thayyil, P. Montaldo, D. Jenkins, G. Quintás, C. Oger, [86] K.B. Pandey, N. Mishra, S.I. Rizvi, Protein oxidation biomarkers in plasma of
J.M. Galano, C. Vigor, T. Durand, M. Vento, J. Kuligowski, Novel free-radical type 2 diabetic patients, Clin. Biochem. 43 (2010) 508–511, https://doi.org/10.
mediated lipid peroxidation biomarkers in newborn plasma, Anal. Chim. Acta 1016/j.clinbiochem.2009.11.011
996 (2017) 88–97, https://doi.org/10.1016/j.aca.2017.09.026 [87] A. Taylor, K.J.A. Davies, Protein oxidation and loss of protease activity may lead
[62] Y. Yoshida, A. Umeno, M. Shichiri, Lipid peroxidation biomarkers for evaluating to cataract formation in the aged lens, Free Radic. Biol. Med. 3 (1987) 371–377,
oxidative stress and assessing antioxidant capacity in vivo, J. Clin. Biochem. https://doi.org/10.1016/0891-5849(87)90015-3
Nutr. 52 (2013) 9–16, https://doi.org/10.3164/jcbn.12-112 [88] E.R. Stadtman, Oxidation of free amino acids and amino acid residues in pro­
[63] J.A. Voynow, A. Kummarapurugu, Isoprostanes and asthma, Biochim. Biophys. teins by radiolysis and by metal-catalyzed reactions, Annu. Rev. Biochem. 62
Acta - Gen. Subj. 1810 (2011) 1091–1095, https://doi.org/10.1016/j.bbagen.2011. (1993) 797–821, https://doi.org/10.1146/annurev.bi.62.070193.004053
04.016 [89] R. Kehm, T. Baldensperger, J. Raupbach, A. Höhn, Protein oxidation - Formation
[64] E. Miller, M. Mrowicka, J. Saluk-Juszczak, M. Ireneusz, The level of isoprostanes mechanisms, detection and relevance as biomarkers in human diseases, Redox
as a non-invasive marker for in vivo lipid peroxidation in secondary progressive Biol. 42 (2021) 101901, https://doi.org/10.1016/j.redox.2021.101901
multiple sclerosis, Neurochem. Res. 36 (2011) 1012–1016, https://doi.org/10. [90] E.R. Stadtman, Protein oxidation and aging, Free Radic. Res. 40 (2006)
1007/s11064-011-0442-1 1250–1258, https://doi.org/10.1080/10715760600918142
[65] C. Peña-Bautista, C. Vigor, J.M. Galano, C. Oger, T. Durand, I. Ferrer, A. Cuevas, [91] E.R. Stadtman, R.L. Levine, Protein oxidation, Ann. N. Y. Acad. Sci. 899 (2000)
R. López-Cuevas, M. Baquero, M. López-Nogueroles, M. Vento, D. Hervás, 191–208, https://doi.org/10.1111/j.1749-6632.2000.tb06187.x
A. García-Blanco, C. Cháfer-Pericás, Plasma lipid peroxidation biomarkers [92] G. Aldini, I. Dalle-Donne, R.M. Facino, A. Milzani, M. Carini, Intervention stra­
for early and non-invasive Alzheimer Disease detection, Free Radic. Biol. tegies to inhibit protein carbonylation by lipoxidation-derived reactive carbo­
Med. 124 (2018) 388–394, https://doi.org/10.1016/j.freeradbiomed.2018.06. nyls, Med. Res. Rev. 27 (2007) 817–868, https://doi.org/10.1002/med.20073
038 [93] F. Alzaid, V.B. Patel, V.R. Preedy, Biomarkers of oxidative stress in blood, in: V.R.
[66] G. Casadesus, M.A. Smith, S. Basu, J. Hua, D.E. Capobianco, S.L. Siedlak, X. Zhu, Preedy, V.B. Patel (Eds.), General Methods in Biomarker Research and Their
G. Perry, Increased isoprostane and prostaglandin are prominent in neurons in Applications, Springer International Publishing, 2015: pp. 567–594. https://doi.
Alzheimer disease, Mol. Neurodegener. 2 (2007) 1–8, https://doi.org/10.1186/ org/10.1007/978–94-007–7696-8_41.
1750-1326-2-2 [94] C.P. Gonzalez-Hunt, M. Wadhwa, L.H. Sanders, DNA damage by oxidative stress:
[67] M. Roest, H.A.M. Voorbij, Y.T. Van der Schouw, P.H.M. Peeters, T. Teerlink, Measurement strategies for two genomes, Curr. Opin. Toxicol. 7 (2018) 87–94,
P.G. Scheffer, High levels of urinary F2-isoprostanes predict cardiovascular https://doi.org/10.1016/j.cotox.2017.11.001
mortality in postmenopausal women, J. Clin. Lipido 2 (2008) 298–303, https:// [95] C. Guo, X. Li, R. Wang, J. Yu, M. Ye, L. Mao, S. Zhang, S. Zheng, Association
doi.org/10.1016/J.JACL.2008.06.004 between oxidative DNA damage and risk of colorectal cancer: Sensitive de­
[68] D. Milatovic, S. Zaja-Milatovic, R.C. Gupta, Biomarkers of oxidative/nitrosative termination of urinary 8-hydroxy-2′-deoxyguanosine by UPLC-MS/MS analysis,
stress and neurotoxicity, in: R.C. Gupta (Ed.), Biomarkers in Toxicology, Sci. Rep. 6 (2016) 32581, https://doi.org/10.1038/srep32581
Academic Press, Boston, 2014, pp. 863–881, https://doi.org/10.1016/B978-0-12- [96] M. Dizdaroglu, P. Jaruga, M. Birincioglu, H. Rodriguez, Free radical-induced
404630-6.00052-X damage to DNA: Mechanisms and measurement, Free Radic. Biol. Med. 32
[69] L.J. Roberts, J.P. Fessel, The biochemistry of the isoprostane, neuroprostane, and (2002) 1102–1115, https://doi.org/10.1016/S0891-5849(02)00826-2
isofuran pathways of lipid peroxidation, Chem. Phys. Lipids 128 (2004) [97] K. Kino, M. Hirao-Suzuki, M. Morikawa, A. Sakaga, H. Miyazawa, Generation,
173–186, https://doi.org/10.1016/J.CHEMPHYSLIP.2003.09.016 repair and replication of guanine oxidation products, Genes Environ. 39 (2017)
[70] W.L. Song, J.A. Lawson, D. Reilly, J. Rokach, C.T. Chang, B. Giasson, 21, https://doi.org/10.1186/s41021-017-0081-0
G.A. FitzGerald, Neurofurans, novel indices of oxidant stress derived from [98] C. Mingard, J. Wu, M. McKeague, S.J. Sturla, Next-generation DNA damage se­
docosahexaenoic acid, J. Biol. Chem. 283 (2008) 6–16, https://doi.org/10.1074/ quencing, Chem. Soc. Rev. 49 (2020) 7354–7377, https://doi.org/10.1039/
JBC.M706124200 D0CS00647E
[71] F. Guéraud, M. Atalay, N. Bresgen, A. Cipak, P.M. Eckl, L. Huc, I. Jouanin, [99] M. Dizdaroglu, P. Jaruga, Mechanisms of free radical-induced damage to DNA,
W. Siems, K. Uchida, Chemistry and biochemistry of lipid peroxidation pro­ Free Radic. Res. 46 (2012) 382–419, https://doi.org/10.3109/10715762.2011.
ducts, Free Radic. Res. 44 (2010) 1098–1124, https://doi.org/10.3109/10715762. 653969
2010.498477 [100] M.S. Cooke, M.D. Evans, M. Dizdaroglu, J. Lunec, Oxidative DNA damage: me­
[72] L.J. Marnett, Oxy radicals, lipid peroxidation and DNA damage, Toxicology chanisms, mutation, and disease, FASEB J. 17 (2003) 1195–1214, https://doi.org/
181–182 (2002) 219–222, https://doi.org/10.1016/S0300-483X(02)00448-1 10.1096/fj.02-0752rev
[73] G. Noberasco, P. Odetti, D. Boeri, M. Maiello, L. Adezati, Malondialdehyde (MDA) [101] B. Halliwell, Antioxidant characterization. Methodology and mechanism,
level in diabetic subjects. Relationship with blood glucose and glycosylated Biochem. Pharm. 49 (1995) 1341–1348, https://doi.org/10.1016/0006-2952(95)
hemoglobin, Biomed. Pharm. 45 (1991) 193–196, https://doi.org/10.1016/0753- 00088-H
3322(91)90107-5 [102] F. Shahidi, Antioxidants: principles and applications, in: F. Shahidi (Ed.),
[74] S. Pandey, B. Singh, S.K. Yadav, A.A. Mahdi, Novel biomarker for neurodegen­ Handbook of Antioxidants for Food Preservation, Woodhead Publishing,
erative diseases- motor neuron disease (MND), cerebellar ataxia (CA) and Amsterdam, 2015, pp. 1–12.
Parkinson’s disease (PD), Clin, Chim. Acta 485 (2018) 258–261, https://doi.org/ [103] D.V. Ratnam, D.D. Ankola, V. Bhardwaj, D.K. Sahana, M.N.V.R. Kumar, Role of
10.1016/j.cca.2018.07.021 antioxidants in prophylaxis and therapy: A pharmaceutical perspective, J.
[75] H. Zhong, H. Yin, Role of lipid peroxidation derived 4-hydroxynonenal (4-HNE) Control. Release 113 (2006) 189–207, https://doi.org/10.1016/j.jconrel.2006.04.
in cancer: Focusing on mitochondria, Redox Biol. 4 (2015) 193–199, https://doi. 015
org/10.1016/j.redox.2014.12.011 [104] Z. Haida, M. Hakiman, A comprehensive review on the determination of en­
[76] G. Poli, R.J. Schaur, W.G. Siems, G. Leonarduzzi, 4-Hydroxynonenal: A mem­ zymatic assay and nonenzymatic antioxidant activities, Food Sci. Nutr. 7 (2019)
brane lipid oxidation product of medicinal interest, Med. Res. Rev. 28 (2008) 1555–1563, https://doi.org/10.1002/fsn3.1012
569–631, https://doi.org/10.1002/med.20117 [105] M.C. Houston, Nutraceuticals, vitamins, antioxidants, and minerals in the pre­
[77] A. Gęgotek, E. Skrzydlewska, Biological effect of protein modifications by lipid vention and treatment of hypertension, Prog. Cardiovasc. Dis. 47 (2005)
peroxidation products, Chem. Phys. Lipids 221 (2019) 46–52, https://doi.org/10. 396–449, https://doi.org/10.1016/j.pcad.2005.01.004
1016/j.chemphyslip.2019.03.011 [106] S.S. Gill, N. Tuteja, Reactive oxygen species and antioxidant machinery in
[78] A.W. Girotti, W. Korytowski, Cholesterol as a natural probe for free radical- abiotic stress tolerance in crop plants, Plant Physiol. Biochem. 48 (2010)
mediated lipid peroxidation in biological membranes and lipoproteins, J. 909–930, https://doi.org/10.1016/j.plaphy.2010.08.016
Chromatogr. B Anal. Technol. Biomed. Life Sci. 1019 (2016) 202–209, https://doi. [107] P. Chelikani, I. Fita, P.C. Loewen, Diversity of structures and properties among
org/10.1016/j.jchromb.2015.12.034 catalases, Cell. Mol. Life Sci. 61 (2004) 192–208, https://doi.org/10.1007/
[79] S. Gill, R. Chow, A.J. Brown, Sterol regulators of cholesterol homeostasis and s00018-003-3206-5
beyond: The oxysterol hypothesis revisited and revised, Prog. Lipid Res. 47 [108] K. Hamre, R. Christiansen, R. Waagbø, A. Maage, B.E. Torstensen, B. Lygren,
(2008) 391–404, https://doi.org/10.1016/j.plipres.2008.04.002 E. Lie, S. Wathne, Albrektsen, Antioxidant vitamins, minerals and lipid levels in
[80] C.L. Hawkins, P.E. Morgan, M.J. Davies, Quantification of protein modification by diets for Atlantic salmon (Salmo salar, L.): Effects on growth performance and
oxidants, Free Radic. Biol. Med. 46 (2009) 965–988, https://doi.org/10.1016/j. fillet quality, Aquac. Nutr. 10 (2004) 113–123, https://doi.org/10.1111/j.1365-
freeradbiomed.2009.01.007 2095.2003.00288.x

23
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

[109] D.W. Reif, Ferritin as a source of iron for oxidative damage, Free Radic. Biol. J. Agric. Food Chem. 64 (2016) 1046–1070, https://doi.org/10.1021/acs.jafc.
Med. 12 (1992) 417–427, https://doi.org/10.1016/0891-5849(92)90091-T 5b04744
[110] T. Byers, G. Perry, Dietary carotenes, vitamin C, and vitamin E as protective [137] A.E. Holley, K.H. Cheeseman, Measuring free radical reactions in vivo, Br. Med.
antioxidants in human cancers, Annu. Rev. Nutr. 12 (1992) 139–159, https://doi. Bull. 49 (1993) 494–505, https://doi.org/10.1093/oxfordjournals.bmb.a072626
org/10.1146/annurev.nu.12.070192.001035 [138] C.M. Spickett, I. Wiswedel, W. Siems, K. Zarkovic, N. Zarkovic, Advances in
[111] M.R. McCall, B. Frei, Can antioxidant vitamins materially reduce oxidative da­ methods for the determination of biologically relevant lipid peroxidation
mage in humans? Free Radic. Biol. Med. 26 (1999) 1034–1053, https://doi.org/ products, Free Radic. Res. 44 (2010) 1172–1202, https://doi.org/10.3109/
10.1016/S0891-5849(98)00302-5 10715762.2010.498476
[112] K.K. Namitha, P.S. Negi, Chemistry and biotechnology of carotenoids, Crit. Rev. [139] B. Halliwell, M. Whiteman, Measuring reactive species and oxidative damage in
Food Sci. Nutr. 50 (2010) 728–760, https://doi.org/10.1080/10408398.2010. vivo and in cell culture: How should you do it and what do the results mean?
499811 Br. J. Pharm. 142 (2004) 231–255, https://doi.org/10.1038/sj.bjp.0705776
[113] G. Britton, Functions of Intact Carotenoids, in: G. Britton, S. Liaaen-Jensen, [140] E. Niki, Antioxidant capacity: Which capacity and how to assess it? J. Berry Res.
H. Pfander (Eds.), Carotenoids, Birkhäuser Basel, 2008, pp. 189–212, https://doi. 1 (2011) 169–176, https://doi.org/10.3233/JBR-2011-018
org/10.1007/978-3-7643-7499-0_10 [141] W. Sattler, D. Mohr, R. Stocker, Rapid isolation of lipoproteins and assessment of
[114] N.I. Krinsky, E.J. Johnson, Carotenoid actions and their relation to health and their peroxidation by high-performance liquid chromatography postcolumn
disease, Mol. Asp. Med. 26 (2005) 459–516, https://doi.org/10.1016/j.mam.2005. chemiluminescence, Methods Enzym. 233 (1994) 469–489, https://doi.org/10.
10.001 1016/S0076-6879(94)33053-0
[115] S. Vertuani, A. Angusti, S. Manfredini, The antioxidants and pro-antioxidants [142] T. Asakawa, S. Matsushita, Colorimetric determination of peroxide value with
network: An overview, Curr. Pharm. Des. 10 (2004) 1677–1694, https://doi.org/ potassium iodide-silica gel reagent, J. Am. Oil Chem. Soc. 1978 558 55 (1978)
10.2174/1381612043384655 619–620, https://doi.org/10.1007/BF02678958
[116] N.I. Krinsky, J.T. Landrum, R.A. Bone, Biologic mechanisms of the protective role [143] S. Hara, Y. Totani, A highly sensitive method for the micro-determination of
of lutein and zeaxanthin in the eye, Annu. Rev. Nutr. 23 (2003) 171–201, https:// lipid hydroperoxides by potentiometry, J. Am. Oil Chem. Soc. 65 (1988)
doi.org/10.1146/annurev.nutr.23.011702.073307 1948–1950, https://doi.org/10.1007/BF02546014
[117] P. Schuler, Natural Antioxidants Exploited Commercially, in: B.J.F. Hudson (Ed.), [144] M. Sano, R. Yoshida, M. Degawa, T. Miyase, Y. Kyoji, Determination of peroxyl
Food Antioxidants, Springer, Netherlands, Dordrecht, 1990, pp. 99–170, https:// radical scavenging activity of flavonoids and plant extracts using an automatic
doi.org/10.1007/978-94-009-0753-9_4 potentiometric titrator, J. Agric. Food Chem. 51 (2003) 2912–2916, https://doi.
[118] P. Molz, N. Schröder, Potential therapeutic effects of lipoic acid on memory org/10.1021/JF0211276
deficits related to aging and neurodegeneration, Front. Pharmacol. 8 (2017) 849, [145] S.E. Çelik, B. Bekdeşer, R. Apak, A novel colorimetric sensor for measuring hy­
https://doi.org/10.3389/FPHAR.2017.00849 droperoxide content and peroxyl radical scavenging activity using starch-sta­
[119] S. Rizvi, S.T. Raza, F. Ahmed, A. Ahmad, S. Abbas, F. Mahdi, The role of Vitamin E bilized gold nanoparticles, Talanta 196 (2019) 32–38, https://doi.org/10.1016/J.
in human health and some diseases, Sultan Qaboos Univ, Med. J. 14 (2014) TALANTA.2018.12.022
157–165 /pmc/articles/PMC3997530/. [146] C.H. Lea, Methods for determining peroxide in lipids, J. Sci. Food Agric. 3 (1952)
[120] W. Rungratanawanich, M. Memo, D. Uberti, Redox homeostasis and natural 586–594, https://doi.org/10.1002/JSFA.2740031205
dietary compounds: Focusing on antioxidants of rice (Oryza sativa L.), Nutrients [147] J.M. DeLong, R.K. Prange, D.M. Hodges, C.F. Forney, M.C. Bishop, M. Quillia,
10 (2018) 1605, https://doi.org/10.3390/NU10111605 Using a modified Ferrous Oxidation−Xylenol Orange (FOX) assay for detection
[121] S. Qiu, Y. Zhao, S. Liu, X. Li, L. Guo, L. Hai, Y. Wu, Design, synthesis and eva­ of lipid hydroperoxides in plant tissue, J. Agric. Food Chem. 50 (2001) 248–254,
luation of dual-targeting prodrug co-modified by organic amine and L-ascorbic https://doi.org/10.1021/JF0106695
acid for CNS delivery, Lett. Drug Des. Discov. 14 (2017) 1065–1072, https://doi. [148] Z.Y. Jiang, J.V. Hunt, S.P. Wolff, Ferrous ion oxidation in the presence of xy­
org/10.2174/1570180814666161230161152 lenol orange for detection of lipid hydroperoxide in low density lipoprotein,
[122] W. Xiao, Q. Fu, Y. Zhao, L. Zhang, Q. Yue, L. Hai, L. Guo, Y. Wu, Ascorbic acid- Anal. Biochem. 202 (1992) 384–389, https://doi.org/10.1016/0003-2697(92)
modified brain-specific liposomes drug delivery system with “lock-in” function, 90122-N
Chem. Phys. Lipids 224 (2019) 104727, https://doi.org/10.1016/J.CHEMPHYSLIP. [149] M. Antolovich, P.D. Prenzler, E. Patsalides, S. McDonald, K. Robards, Methods for
2019.01.005 testing antioxidant activity, Analyst 127 (2002) 183–198, https://doi.org/10.
[123] R. Apak, K. Güçlü, B. Demirata, M. Özyürek, S.E. Çelik, B. Bektaşoǧlu, K.I. Berker, 1039/B009171P
D. Özyurt, Comparative evaluation of various total antioxidant capacity assays [150] W.A. Pryor, J.A. Cornicelli, L.J. Devall, B. Tait, B.K. Trivedi, D.T. Witiak, M. Wu, A
applied to phenolic compounds with the CUPRAC assay, Molecules 12 (2007) rapid screening test to determine the antioxidant potencies of natural and
1496–1547, https://doi.org/10.3390/12071496 synthetic antioxidants, J. Org. Chem. 58 (1993) 3521–3532, https://doi.org/10.
[124] C.A. Rice-Evans, N.J. Miller, G. Paganga, Structure-antioxidant activity re­ 1021/JO00065A013
lationships of flavonoids and phenolic acids, Free Radic. Biol. Med. 20 (1996) [151] I.M. Heinonen, P.J. Lehtonen, A.I. Hopia, Antioxidant activity of berry and fruit
933–956, https://doi.org/10.1016/0891-5849(95)02227-9 wines and liquors, J. Agric. Food Chem. 46 (1998) 25–31, https://doi.org/10.
[125] H.J.H. Fenton, Oxidation of tartaric acid in presence of iron, J. Chem. Soc. Trans. 1021/JF970489O
65 (1894) 899–910, https://doi.org/10.1039/CT8946500899 [152] N. Adhoum, L. Monser, Electrochemical sensor for hydroperoxides determina­
[127] P.C.H. Hollman, M.B. Katan, Dietary flavonoids: Intake, health effects and tion based on Prussian blue film modified electrode, Sens. Actuators B Chem.
bioavailability, Food Chem. Toxicol. 37 (1999) 937–942, https://doi.org/10.1016/ 133 (2008) 588–592, https://doi.org/10.1016/J.SNB.2008.03.039
S0278-6915(99)00079-4 [153] J.M.C. Gutteridge, B. Halliwell, The measurement and mechanism of lipid per­
[128] S.A. Aherne, N.M. O’Brien, Dietary flavonols: Chemistry, food content, and oxidation in biological systems, Trends Biochem. Sci. 15 (1990) 129–135,
metabolism, Nutrition 18 (2002) 75–81, https://doi.org/10.1016/S0899- https://doi.org/10.1016/0968-0004(90)90206-Q
9007(01)00695-5 [154] R. Amorati, L. Valgimigli, Advantages and limitations of common testing
[129] J.E.N. Dolatabadi, S. Kashanian, A review on DNA interaction with synthetic methods for antioxidants, Free Radic. Res. 49 (2015) 633–649, https://doi.org/
phenolic food additives, Food Res. Int. 43 (2010) 1223–1230, https://doi.org/10. 10.3109/10715762.2014.996146
1016/j.foodres.2010.03.026 [155] R. Guillén-Sans, M. Guzmán-Chozas, The Thiobarbituric Acid (TBA) reaction in
[130] A. Ghiselli, M. Serafini, F. Natella, C. Scaccini, Total antioxidant capacity as a foods: A Review, Crit. Rev. Food Sci. Nutr. 38 (1998) 315–350, https://doi.org/10.
tool to assess redox status: Critical view and experimental data, Free Radic. 1080/10408699891274228
Biol. Med. 29 (2000) 1106–1114, https://doi.org/10.1016/S0891-5849(00) [156] D. Jardine, M. Antolovich, P.D. Prenzler, K. Robards, Liquid chromato­
00394-4 graphy−mass spectrometry (LC-MS) investigation of the Thiobarbituric Acid
[131] P. Jackson, C.M. Loughrey, J.H. Lightbody, P.T. Mcnamee, I.S. Young, Effect of Reactive Substances (TBARS) reaction, J. Agric. Food Chem. 50 (2002)
hemodialysis on total antioxidant capacity and serum antioxidants in patients 1720–1724, https://doi.org/10.1021/JF011336A
with chronic renal failure, Clin. Chem. 41 (1995) 1138. [157] T. Bakır, B.Y. Beker, İ. Sönmezoğlu, F. İmer, R. Apak, Antioxidant and prooxidant
[132] R.L. Prior, G. Cao, In vivo total antioxidant capacity: Comparison of different effects of α-tocopherol in a linoleic acid-copper(II)-ascorbate system, Eur. J.
analytical methods, Free Radic. Biol. Med. 27 (1999) 1173–1181, https://doi.org/ Lipid Sci. Technol. 115 (2013) 372–376, https://doi.org/10.1002/EJLT.201200124
10.1016/S0891-5849(99)00203-8 [158] J.R. Seal, C.M. Havrilla, N.A. Porter, D.L. Hachey, Analysis of unsaturated com­
[133] R. Apak, M. Özyürek, K. Güçlü, E. Çapanoğlu, Antioxidant activity/capacity pounds by Ag+ coordination ionspray mass spectrometry: studies of the for­
measurement. 2. Hydrogen Atom Transfer (HAT)-based, mixed-mode (Electron mation of the Ag+/lipid complex, J. Am. Soc. Mass Spectrom. 14 (2003)
Transfer (ET)/HAT), and lipid peroxidation assays, J. Agric. Food Chem. 64 (2016) 872–880, https://doi.org/10.1016/S1044-0305(03)00339-8
1028–1045, https://doi.org/10.1021/ACS.JAFC.5B04743 [159] Y. Kawai, S. Takeda, T. Junji, Lipidomic analysis for lipid peroxidation-derived
[134] S. Uzunboy, S. Demirci Çekiç, R. Apak, Determination of cobalt(II)-hydrogen aldehydes using gas chromatography-mass spectrometry, Chem. Res. Toxicol.
peroxide-induced DNA oxidative damage and preventive antioxidant activity 20 (2007) 99–107, https://doi.org/10.1021/TX060199E
by CUPRAC colorimetry, Anal. Lett. 52 (2019) 2663–2676, https://doi.org/10. [160] F. Tietze, Enzymic method for quantitative determination of nanogram
1080/00032719.2019.1587448 amounts of total and oxidized glutathione: Applications to mammalian blood
[135] S. Uzunboy, S.D. Çekiç, E. Eksin, A. Erdem, R. Apak, CUPRAC colorimetric and and other tissues, Anal. Biochem. 27 (1969) 502–522, https://doi.org/10.1016/
electroanalytical methods determining antioxidant activity based on preven­ 0003-2697(69)90064-5
tion of oxidative DNA damage, Anal. Biochem. 518 (2017) 69–77, https://doi. [161] O. Erel, S. Neselioglu, A novel and automated assay for thiol/disulphide
org/10.1016/j.ab.2016.10.028 homeostasis, Clin. Biochem. 47 (2014) 326–332, https://doi.org/10.1016/J.
[136] R. Apak, M. Özyürek, K. Güçlü, E. Çapanoğlu, Antioxidant activity/capacity CLINBIOCHEM.2014.09.026
measurement. 3. Reactive oxygen and nitrogen species (ROS/RNS) scavenging [162] E.M. Görüşük, B. Bekdeşer, M. Bener, R. Apak, ABTS radical-based single reagent
assays, oxidative stress biomarkers, and chromatographic/chemometric assays, assay for simultaneous determination of biologically important thiols and

24
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

disulfides, Talanta 218 (2020) 121212, https://doi.org/10.1016/J.TALANTA.2020. treatment and bone metastasis, Biomed. Pharmacother. 65 (2011) 516–524,
121212 https://doi.org/10.1016/j.biopha.2011.06.003
[163] A. Çifteci, S.E. Çelik, R. Apak, Gold–nanoparticle based turn–on fluorometric [185] S. Shukla, J.K. Srivastava, E. Shankar, R. Kanwal, A. Nawab, H. Sharma,
sensor for quantification of sulfhydryl and disulfide forms of biothiols: mea­ N. Bhaskaran, L.E. Ponsky, P. Fu, G.T. MacLennan, S. Gupta, Oxidative stress and
surement of thiol/disulfide homeostasis, Anal. Lett. (2021) (accepted manu­ antioxidant status in high-risk prostate cancer subjects, Diagnostics 10 (2020)
script). 126, https://doi.org/10.3390/diagnostics10030126
[164] K.D. Jacob, N. Noren Hooten, A.R. Trzeciak, M.K. Evans, Markers of oxidant stress [186] F.F. Chiang, H.M. Wang, Y.C. Lan, M.H. Yang, S.C. Huang, Y.C. Huang, High
that are clinically relevant in aging and age-related disease, Mech. Ageing Dev. homocysteine is associated with increased risk of colorectal cancer in­
134 (2013) 139–157, https://doi.org/10.1016/J.MAD.2013.02.008 dependently of oxidative stress and antioxidant capacities, Clin. Nutr. 33 (2014)
[165] R.L. Levine, D. Garland, C.N. Oliver, A. Amici, I. Climent, A.G. Lenz, B.W. Ahn, 1054–1060, https://doi.org/10.1016/j.clnu.2013.11.007
S. Shaltiel, E.R. Stadtman, Determination of carbonyl content in oxidatively [187] J. Zińczuk, M. Maciejczyk, K. Zaręba, A. Pryczynicz, V. Dymicka-Piekarska,
modified proteins, Methods Enzym. 186 (1990) 464–478, https://doi.org/10. J. Kamińska, O. Koper-Lenkiewicz, J. Matowicka-Karna, B. Kędra, A. Zalewska,
1016/0076-6879(90)86141-H K. Guzińska-Ustymowicz, Pro-oxidant enzymes, redox balance and oxidative
[166] N. Purdel, Current methods used in the protein carbonyl assay, Annu. Res. Rev. damage to proteins, lipids and DNA in colorectal cancer tissue. Is oxidative
Biol. 4 (2014) 2015–2026, https://doi.org/10.9734/ARRB/2014/8763 stress dependent on tumour budding and inflammatory infiltration? Cancers
[167] G. Bayarsaikhan, A.N. Avan, S.D. Çekiç, R. Apak, Use of modified CUPRAC and 12 Basel, 2020, p. 1636, https://doi.org/10.3390/cancers12061636
dinitrophenylhydrazine colorimetric methods for simultaneous measurement [188] A. Shrivastava, S.P. Mishra, S. Pradhan, S. Choudhary, S. Singla, K. Zahra,
of oxidative protein damage and antioxidant defense against oxidation, Talanta L.M. Aggarwal, An assessment of serum oxidative stress and antioxidant
204 (2019) 613–625, https://doi.org/10.1016/J.TALANTA.2019.06.049 parameters in patients undergoing treatment for cervical cancer, Free Radic.
[168] C.S. Mesquita, R. Oliveira, F. Bento, D. Geraldo, J.V. Rodrigues, J.C. Marcos, Biol. Med. 167 (2021) 29–35, https://doi.org/10.1016/j.freeradbiomed.2021.02.
Simplified 2,4-dinitrophenylhydrazine spectrophotometric assay for quantifi­ 037
cation of carbonyls in oxidized proteins, Anal. Biochem. 458 (2014) 69–71, [189] V. Battisti, L.D.K. Maders, M.D. Bagatini, K.F. Santos, R.M. Spanevello,
https://doi.org/10.1016/J.AB.2014.04.034 P.A. Maldonado, A.O. Brulé, M. do, C. Araújo, M.R.C. Schetinger, V.M. Morsch,
[169] H. Ma, S. Liu, N. Zheng, Y. Liu, X.X. Han, C. He, H. Lu, B. Zhao, Frequency shifts in Measurement of oxidative stress and antioxidant status in acute lymphoblastic
surface-enhanced raman spectroscopy-based immunoassays: mechanistic in­ leukemia patients, Clin. Biochem. 41 (2008) 511–518, https://doi.org/10.1016/j.
sights and application in protein carbonylation detection, Anal. Chem. 91 (2019) clinbiochem.2008.01.027
9376–9381, https://doi.org/10.1021/ACS.ANALCHEM.9B02640 [190] H. Blasco, G. Garcon, F. Patin, C. Veyrat-Durebex, J. Boyer, D. Devos, P. Vourc’H,
[170] T.A. Enache, E. Matei, V.C. Diculescu, Electrochemical sensor for carbonyl groups C.R. Andres, P. Corcia, Panel of oxidative stress and inflammatory biomarkers in
in oxidized proteins, Anal. Chem. 91 (2019) 1920–1927, https://doi.org/10.1021/ ALS: A pilot study, Can. J. Neurol. Sci. 44 (2017) 90–95, https://doi.org/10.1017/
ACS.ANALCHEM.8B03969 cjn.2016.284
[171] B. Sun, Y. Gou, Z. Xue, X. Zheng, Y. Ma, F. Hu, W. Zhao, Protections of bovine [191] C.C. de Farias, M. Maes, K.L. Bonifácio, C.C. Bortolasci, A. de Souza Nogueira,
serum albumin protein from damage on functionalized graphene-based elec­ F.F. Brinholi, A.K. Matsumoto, M.A. do Nascimento, L.B. de Melo, S.L. Nixdorf,
trodes by flavonoids, Mater. Sci. Eng. C. 62 (2016) 197–205, https://doi.org/10. E.L. Lavado, E.G. Moreira, D.S. Barbosa, Highly specific changes in antioxidant
1016/J.MSEC.2016.01.043 levels and lipid peroxidation in Parkinson’s disease and its progression: Disease
[172] B. Halliwell, Why and how should we measure oxidative DNA damage in nu­ and staging biomarkers and new drug targets, Neurosci. Lett. 617 (2016) 66–71,
tritional studies? How far have we come? Am. J. Clin. Nutr. 72 (2000) https://doi.org/10.1016/j.neulet.2016.02.011
1082–1087, https://doi.org/10.1093/AJCN/72.5.1082 [192] J. Miletić, D. Drakulić, S. Pejić, M. Petković, T.V. Ilić, M. Miljković, A. Stefanović,
[173] B. Halliwell, Can oxidative DNA damage be used as a biomarker of cancer risk in M. Prostran, M. Stojanov, Prooxidant–antioxidant balance, advanced oxidation
humans? Problems, resolutions and preliminary results from nutritional sup­ protein products and lipid peroxidation in Serbian patients with Parkinson’s
plementation studies, Free Radic. Res. 29 (1998) 469–486, https://doi.org/10. disease, Int. J. Neurosci. 128 (2018) 600–607, https://doi.org/10.1080/00207454.
1080/10715769800300531 2017.1403916
[174] S. Senturker, M. Dizdaroglu, The effect of experimental conditions on the levels [193] M.S. Medeiros, A. Schumacher-Schuh, A.M. Cardoso, G.V. Bochi, J. Baldissarelli,
of oxidatively modified bases in DNA as measured by gas chromatography-mass A. Kegler, D. Santana, C.M.M.B.S. Chaves, M.R.C. Schetinger, R.N. Moresco,
spectrometry: How many modified bases are involved? Prepurification or not? C.R.M. Rieder, M.R. Fighera, Iron and oxidative stress in Parkinson’s disease: An
Free Radic. Biol. Med. 27 (1999) 370–380, https://doi.org/10.1016/S0891- observational study of injury biomarkers, PLoS One 11 (2016) e0146129,
5849(99)00069-6 https://doi.org/10.1371/journal.pone.0146129
[175] B. Halliwell, A. Adhikary, M. Dingfelder, M. Dizdaroglu, Hydroxyl radical is a [194] H. Negahdar, S.R. Hosseini, H. Parsian, F. Kheirkhah, A. Mosapour, S. Khafri,
significant player in oxidative DNA damage in vivo, Chem. Soc. Rev. (2021), A.H. Haghighi, Homocysteine, trace elements and oxidant/antioxidant
https://doi.org/10.1039/D1CS00044F status in mild cognitively impaired elderly persons: a cross-sectional study,
[176] A.-M. Chiorcea-Paquim, A.M. Oliveira-Brett, Nanostructured material-based Rom. J. Intern. Med. 53 (2015) 336–342, https://doi.org/10.1515/rjim-2015-
electrochemical sensing of oxidative DNA damage biomarkers 8-oxoguanine 0043
and 8-oxodeoxyguanosine: a comprehensive review, Mikrochim. Acta 188 [195] O.M.E. Abdel-Salam, E.R. Youness, N.A. Mohammed, W.A.A. Elhamed, Nuclear
(2021) 58, https://doi.org/10.1007/S00604-020-04689-7 Factor-Kappa B and Other Oxidative Stress Biomarkers in Serum of Autistic
[177] S. Dawbaa, Ö. Aybastıer, C. Demir, Ultrasensitive determination of DNA oxida­ Children, Open, J. Mol. Integr. Physiol. 05 (2015) 18–27, https://doi.org/10.4236/
tion products by gas chromatography–tandem mass spectrometry and the role ojmip.2015.51002
of antioxidants in the prevention of oxidative damage, J. Chromatogr. B. 1051 [196] R. Alghazeer, N. Alghazir, N. Awayn, O. Ahtiwesh, S. Elgahmasi, Biomarkers of
(2017) 84–91, https://doi.org/10.1016/J.JCHROMB.2017.03.014 oxidative stress and antioxidant defense in patients with type 1 diabetes
[178] Q. Xu, H. Yuan, X. Dong, Y. Zhang, M. Asif, Z. Dong, W. He, J. Ren, Y. Sun, F. Xiao, mellitus, Ibnosina J. Med, Biomed. Sci. 10 (2018) 204, https://doi.org/10.4103/
Dual nanoenzyme modified microelectrode based on carbon fiber coated with ijmbs.ijmbs_59_18
AuPd alloy nanoparticles decorated graphene quantum dots assembly for [197] P. Codoñer-Franch, S. Pons-Morales, L. Boix-García, V. Valls-Bellés, Oxidant/
electrochemical detection in clinic cancer samples, Biosens. Bioelectron. 107 antioxidant status in obese children compared to pediatric patients with type 1
(2018) 153–162, https://doi.org/10.1016/J.BIOS.2018.02.026 diabetes mellitus, Pediatr. Diabetes 11 (2010) 251–257, https://doi.org/10.1111/j.
[179] T. Wu, L. Li, G. Song, M. Ran, X. Lu, X. Liu, An ultrasensitive electrochemical 1399-5448.2009.00565.x
sensor based on cotton carbon fiber composites for the determination of su­ [198] B. Salmonowicz, M. Krzystek-Korpacka, A. Noczyńska, Trace elements, magne­
peroxide anion release from cells, Mikrochim. Acta 186 (2019) 198, https://doi. sium, and the efficacy of antioxidant systems in children with Type 1 Diabetes
org/10.1007/S00604-019-3304-1 Mellitus and in their siblings, Adv. Clin. Exp. Med. 23 (2014) 259–268, https://
[180] M.K. Beissenhirtz, F.W. Scheller, F. Lisdat, A superoxide sensor based on a doi.org/10.17219/acem/37074
multilayer cytochrome c electrode, Anal. Chem. 76 (2004) 4665–4671, https:// [199] A. Siddiqui, N.G. Desai, S.B. Sharma, M. Aslam, U.K. Sinha, S.V. Madhu,
doi.org/10.1021/AC049738F Association of oxidative stress and inflammatory markers with chronic stress in
[181] N. Kilic, M. Yavuz Taslipinar, Y. Guney, E. Tekin, E. Onuk, An Investigation into patients with newly diagnosed type 2 diabetes, Diabetes Metab. Res. Rev. 35
the Serum Thioredoxin, Superoxide Dismutase, Malondialdehyde, and (2019) e3147, https://doi.org/10.1002/dmrr.3147
Advanced Oxidation Protein Products in Patients with Breast Cancer, Ann. Surg. [200] R.S. Tupe, A.G. Diwan, V.D. Mittal, P.S. Narayanam, K.B. Mahajan, Association of
Oncol. 21 (2014) 4139–4143, https://doi.org/10.1245/s10434-014-3859-3 plasma proteins at multiple stages of glycation and antioxidant status with
[182] L.K. Reitz, S. de, L. Baptista, E. da, S. Santos, P.F. Hinnig, G. Rockenbach, erythrocyte oxidative stress in patients with type 2 diabetes, Br, J. Biomed. Sci.
F.G.K. Vieira, M.A.A. de Assis, E.L. da Silva, B.C.B. Boaventura, P.F.Di Pietro, Diet 71 (2014) 93–99, https://doi.org/10.1080/09674845.2014.11669971
quality is associated with serum antioxidant capacity in women with breast [201] B. Usluoğullari, C.A. Usluogullari, F. Balkan, M. Orkmez, Role of serum levels of
cancer: A cross sectional study, Nutrients 13 (2021) 1–14, https://doi.org/10. irisin and oxidative stress markers in pregnant women with and without ge­
3390/nu13010115 stational diabetes, Gynecol. Endocrinol. 33 (2017) 405–407, https://doi.org/10.
[183] I.E. Bassey, B.A. Emodi, U.O. Akpan, I.F.A. Iyakndue, E.A. Anakebe, B.E. Icha, 1080/09513590.2017.1284789
H.A. Efobi, A.J. Ntinya, A.E. Udoh, Impact of androgen deprivation on oxidative [202] M. Shang, J. Zhao, L. Yang, L. Lin, Oxidative stress and antioxidant status in
stress and antioxidant status in Nigerian patients with prostate cancer under­ women with gestational diabetes mellitus diagnosed by IADPSG criteria,
going androgen deprivation therapy, JCO Glob. Oncol. 6 (2020) 1481–1489, Diabetes Res. Clin. Pract. 109 (2015) 404–410, https://doi.org/10.1016/j.diabres.
https://doi.org/10.1200/go.20.00290 2015.05.010
[184] V. Battisti, L.D.K. Maders, M.D. Bagatini, L.G.B. Reetz, J. Chiesa, I.E. Battisti, [203] L. Vera-Ramirez, P. Sanchez-Rovira, M.C. Ramirez-Tortosa, C.L. Ramirez-Tortosa,
J.F. Gonçalves, M.M.F. Duarte, M.R.C. Schetinger, V.M. Morsch, Oxidative stress S. Granados-Principal, J.A. Lorente, J.L. Quiles, Free radicals in breast carcino­
and antioxidant status in prostate cancer patients: Relation to Gleason score, genesis, breast cancer progression and cancer stem cells. Biological bases to

25
S. Demirci-Çekiç, G. Özkan, A.N. Avan et al. Journal of Pharmaceutical and Biomedical Analysis 209 (2022) 114477

develop oxidative-based therapies, Crit. Rev. Oncol. Hematol. 80 (2011) [218] A. Burns, M. Zaudig, Mild cognitive impairment in older people, Lancet 360
347–368, https://doi.org/10.1016/j.critrevonc.2011.01.004 (2002) 1963–1965, https://doi.org/10.1016/S0140-6736(02)11920-9
[204] A. Villers, P. Grosclaude, Épidémiologie du cancer de la prostate, Artic. De. Rev., [219] J.N. Constantino, T. Charman, Diagnosis of autism spectrum disorder: re­
Med. Nucl. 32 (2008) 2–4, https://doi.org/10.1016/j.mednuc.2007.11.003 conciling the syndrome, its diverse origins, and variation in expression, Lancet
[205] A. Bhattacharyya, R. Chattopadhyay, S. Mitra, S.E. Crowe, Oxidative stress: An Neurol. 15 (2016) 279–291, https://doi.org/10.1016/S1474-4422(15)00151-9
essential factor in the pathogenesis of gastrointestinal mucosal diseases, [220] P. Steullet, J.H. Cabungcal, J. Coyle, M. Didriksen, K. Gill, A.A. Grace, T.K. Hensch,
Physiol. Rev. 94 (2014) 329–354, https://doi.org/10.1152/physrev.00040.2012 A.S. Lamantia, L. Lindemann, T.M. Maynard, U. Meyer, H. Morishita,
[206] A. Jemal, F. Bray, M.M. Center, J. Ferlay, E. Ward, D. Forman, Global cancer P. O’Donnell, M. Puhl, M. Cuenod, K.Q. Do, Oxidative stress-driven parvalbumin
statistics, Ca. Cancer J. Clin. 61 (2011) 69–90, https://doi.org/10.3322/caac. interneuron impairment as a common mechanism in models of schizophrenia,
20107 Mol. Psychiatry 22 (2017) 936–943, https://doi.org/10.1038/mp.2017.47
[207] A. Ulvik, S.E. Vollset, S. Hansen, R. Gislefoss, E. Jellum, P.M. Ueland, Colorectal [221] P.R. Itankar, S.J. Lokhande, P.R. Verma, S.K. Arora, R.A. Sahu, A.T. Patil,
cancer and the methylenetetrahydrofolate reductase 677C → T and methionine Antidiabetic potential of unripe Carissa carandas Linn. fruit extract, J.
synthase 2756A → G Polymorphisms: A Study of 2,168 case-control pairs from Ethnopharmacol. 135 (2011) 430–433, https://doi.org/10.1016/j.jep.2011.03.036
the JANUS cohort, Cancer Epidemiol. Biomark. Prev. 13 (2004) 2175–2180. [222] L. Guariguata, D.R. Whiting, I. Hambleton, J. Beagley, U. Linnenkamp, J.E. Shaw,
[208] F. Maffei, C. Angeloni, M. Malaguti, J.M.Z. Moraga, F. Pasqui, C. Poli, A. Colecchia, Global estimates of diabetes prevalence for 2013 and projections for 2035,
D. Festi, P. Hrelia, S. Hrelia, Plasma antioxidant enzymes and clastogenic factors Diabetes Res. Clin. Pract. 103 (2014) 137–149, https://doi.org/10.1016/j.diabres.
as possible biomarkers of colorectal cancer risk, Mutat. Res. - Fundam. Mol. 2013.11.002
Mech. Mutagen. 714 (2011) 88–92, https://doi.org/10.1016/j.mrfmmm.2011.06. [223] A. Naudi, M. Jove, V. Ayala, A. Cassanye, J. Serrano, H. Gonzalo, J. Boada, J. Prat,
016 M. Portero-Otin, R. Pamplona, Cellular dysfunction in diabetes as maladaptive
[209] M.H. Stipanuk, Sulfur amino acid metabolism: Pathways for production and response to mitochondrial oxidative stress, Exp. Diabetes Res. 2012 (2012)
removal of homocysteine and cysteine, Annu. Rev. Nutr. 24 (2004) 539–577, 696215, https://doi.org/10.1155/2012/696215
https://doi.org/10.1146/annurev.nutr.24.012003.132418 [224] S. Mariyam, J. Jose, S. Kumar, M.Y. Parmar, A.P.J. Abdul, Super power of anti­
[210] D. Zanini, R. Schmatz, L.P. Pelinson, V.C. Pimentel, P. Da Costa, A.M.H. Cardoso, oxidant in oxidative stress and Diabetes Mellitus, J. Diabetes Endocrinol. Res 2
C.C. Martins, C.C. Schetinger, J. Baldissareli, M. Do Carmo Araújo, L. Oliveira, (2020) 2020.
J. Chiesa, V.M. Morsch, D.B.R. Leal, M.R.C. Schetinger, Ectoenzymes and choli­ [225] H.Z. Pan, L. Zhang, M.Y. Guo, H. Sui, H. Li, W.H. Wu, N.Q. Qu, M.H. Liang,
nesterase activity and biomarkers of oxidative stress in patients with lung D. Chang, The oxidative stress status in diabetes mellitus and diabetic ne­
cancer, Mol. Cell. Biochem. 374 (2013) 137–148, https://doi.org/10.1007/s11010- phropathy, Acta Diabetol. 47 (2010) 71–76, https://doi.org/10.1007/s00592-
012-1513-6 009-0128-1
[211] H.H. Wasserman, J.L. Ives, Reaction of singlet oxygen with enamino carbonyl [226] F.B. Hu, M.J. Stampfer, Is type 2 diabetes mellitus a vascular condition?
systems. A General method for the synthesis of a-keto derivatives of lactones, Arterioscler. Thromb. Vasc. Biol. 23 (2003) 1715–1716, https://doi.org/10.1161/
esters, amides, lactams, and ketones, J. Org. Chem. 50 (1985) 3573–3580, 01.ATV.0000094360.38911.71
https://doi.org/10.1021/jo00219a025 [227] J.L. Evans, I.D. Goldfine, B.A. Maddux, G.M. Grodsky, Are oxidative stress -
[212] P.M. Andersen, Amyotrophic lateral sclerosis associated with mutations in the Activated signaling pathways mediators of insulin resistance and β-cell dys­
CuZn superoxide dismutase gene, Curr. Neurol. Neurosci. Rep. 6 (2006) 37–46, function? Diabetes 52 (2003) 1–8, https://doi.org/10.2337/diabetes.52.1.1
https://doi.org/10.1007/s11910-996-0008-9 [228] M.P. Cohen, F.N. Ziyadeh, S. Chen, Amadori-modified glycated serum proteins
[213] G.N. Babu, A. Kumar, R. Chandra, S.K. Puri, R.L. Singh, J. Kalita, U.K. Misra, and accelerated atherosclerosis in diabetes: Pathogenic and therapeutic im­
Oxidant-antioxidant imbalance in the erythrocytes of sporadic amyotrophic plications, J. Lab. Clin. Med. 147 (2006) 211–219, https://doi.org/10.1016/J.LAB.
lateral sclerosis patients correlates with the progression of disease, Neurochem. 2005.12.006
Int. 52 (2008) 1284–1289, https://doi.org/10.1016/j.neuint.2008.01.009 [229] P. Rondeau, E. Bourdon, The glycation of albumin: Structural and functional im­
[214] G. Anderson, M. Maes, Neurodegeneration in Parkinson’s disease: Interactions pacts, Biochimie 93 (2011) 645–658, https://doi.org/10.1016/j.biochi.2010.12.003
of oxidative stress, tryptophan catabolites and depression with mitochondria [230] M.A. Grillo, S. Colombatto, Advanced glycation end-products (AGEs):
and sirtuins, Mol. Neurobiol. 49 (2014) 771–783, https://doi.org/10.1007/ Involvement in aging and in neurodegenerative diseases, Amino Acids 35
s12035-013-8554-z (2008) 29–36, https://doi.org/10.1007/s00726-007-0606-0
[215] R. Perfeito, T. Cunha-Oliveira, A.C. Rego, Revisiting oxidative stress and mi­ [231] S. Schneider, B. Hoeft, N. Freerksen, B. Fischer, S. Roehrig, S. Yamamoto, H. Maul,
tochondrial dysfunction in the pathogenesis of Parkinson disease - Resemblance to Neonatal complications and risk factors among women with gestational dia­
the effect of amphetamine drugs of abuse, Free Radic. Biol. Med. 53 (2012) betes mellitus, Acta Obstet. Gynecol. Scand. 90 (2011) 231–237, https://doi.org/
1791–1806, https://doi.org/10.1016/j.freeradbiomed.2012.08.569 10.1111/j.1600-0412.2010.01040.x
[216] K.M. Smith, N. Dahodwala, Sex differences in Parkinson’s disease and other [232] R. Gorgal, E. Gonçalves, M. Barros, G. Namora, Â. Magalhães, T. Rodrigues,
movement disorders, Exp. Neurol. 259 (2014) 44–56, https://doi.org/10.1016/j. N. Montenegro, Gestational diabetes mellitus: A risk factor for non-elective
expneurol.2014.03.010 cesarean section, J. Obstet. Gynaecol. Res 38 (2012) 154–159, https://doi.org/10.
[217] I. Pichler, M.F. Del Greco, M. Gögele, C.M. Lill, L. Bertram, C.B. Do, N. Eriksson, 1111/j.1447-0756.2011.01659.x
T. Foroud, R.H. Myers, M. Nalls, M.F. Keller, B. Benyamin, J.B. Whitfield, [233] N. Vrachnis, P. Belitsos, S. Sifakis, K. Dafopoulos, C. Siristatidis, K.I. Pappa,
P.P. Pramstaller, A.A. Hicks, J.R. Thompson, C. Minelli, Serum iron levels and the Z. Iliodromiti, Role of adipokines and other inflammatory mediators in gesta­
risk of Parkinson Disease: A Mendelian Randomization study, PLoS Med 10 tional diabetes mellitus and previous gestational diabetes mellitus, Int. J.
(2013) e1001462, https://doi.org/10.1371/journal.pmed.1001462 Endocrinol. 2012 (2012) 549748, https://doi.org/10.1155/2012/549748

26

You might also like