Sei sulla pagina 1di 10

Analytica Chimica Acta 763 (2013) 1–10

Contents lists available at SciVerse ScienceDirect

Analytica Chimica Acta


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

Review

Evaluating the antioxidant capacity of natural products: A review on


chemical and cellular-based assays
Camilo López-Alarcón a , Ana Denicola b,∗
a
Departamento de Farmacia, Facultad de Química, Pontificia Universidad Católica de Chile, Av. Vicuña Mackenna 4860, 7820436, Santiago, Chile
b
Instituto de Química Biológica, Facultad de Ciencias, Center for Free Radical and Biomedical Research, Universidad de la República, Iguá 4225, 11300
Montevideo, Uruguay

h i g h l i g h t s g r a p h i c a l a b s t r a c t

 There is high interest on determi-


nation of antioxidant capacity of
natural products.
 Several chemical in vitro methods
should be used.
 Cellular antioxidant activity assays
should also be performed.
 No strong correlation between
chemical and cellular assays.

a r t i c l e i n f o a b s t r a c t

Article history: Oxidative stress is associated with several pathologies like cardiovascular, neurodegenerative, cancer and
Received 1 July 2012 even aging. It has been suggested that a diet rich in antioxidants would be beneficial to human health
Received in revised form and a lot of interest is focused on the determination of antioxidant capacity of natural products. Different
24 November 2012
chemical methods have been developed including the popular ORAC that evaluates the potential of a
Accepted 26 November 2012
sample as inhibitor of a target molecule oxidation. Chemical-based methods are useful for screening,
Available online 5 December 2012
they are low cost, high-throughput and yield an index value (expressed as equivalents of Trolox) that
allows comparing and ordering different products. More recently, nanoparticles-based assays have been
Keywords:
Antioxidant capacity
developed to sense the antioxidant power of natural products. However, the antioxidant capacity indexes
Chemical assays obtained by chemical assays cannot extrapolate the performance of the sample in vivo. Considering that
Nanoparticle assays antioxidant action is not limited to scavenging free radicals but includes upregulation of antioxidant and
Cellular assays detoxifying enzymes, modulation of redox cell signaling and gene expression, it is necessary to move to
Reactive oxygen species cellular assays in order to evaluate the potential antioxidant activity of a compound or extract. Animal
Reactive nitrogen species models and human studies are more appropriate but also more expensive and time-consuming, mak-
ing the cell culture assays very attractive as intermediate testing methods. Cellular antioxidant activity
(CAA) assays, activation of redox transcription factors, inhibition of oxidases or activation of antioxidant
enzymes are reviewed and compared with the classical in vitro chemical-based assays for evaluation of
antioxidant capacity of natural products.
© 2012 Elsevier B.V. All rights reserved.

Abbreviations: ROS, reactive oxygen species; RNS, reactive nitrogen species; SOD, superoxide dismutase; DPPH• , 2,2-diphenyl-1-picrylhydrazyl radical; ABTS• +, 2,2 -
azinobis-(3-ethylbenzothiazole-6-sulphonate) radical cation; FRAP, ferric reducing antioxidant power; CUPRAC, cupric ion reducing antioxidant capacity; ORAC, oxygen
radical absorbance capacity; TRAP, total radical-trapping antioxidant parameter; LDL, low density lipoprotein; ESR, electron spin resonance; HepG2, human hepatocarcinoma;
DCFH2 , dihydrodichlorofluorescein; DCFH2 -DA, dihydrodichlorofluorescein diacetate; CAA, cellular antioxidant activity; RBC, red blood cells; NOx, NADPH oxidases; NOS,
nitric oxide synthase; Nrf-2, nuclear factor E2-related protein 2; NF-␬B, nuclear factor kappa B.
∗ Corresponding author. Tel.: +598 2 5250749; fax: +598 2 5250749.
E-mail addresses: clopezr@uc.cl (C. López-Alarcón), denicola@fcien.edu.uy (A. Denicola).

0003-2670/$ – see front matter © 2012 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.aca.2012.11.051
2 C. López-Alarcón, A. Denicola / Analytica Chimica Acta 763 (2013) 1–10

Contents
1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
1.1. Mechanisms of antioxidant actions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
1.1.1. The classic (traditional) antioxidant mechanism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
1.1.2. New mechanisms based on redox signaling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2. Methodologies to evaluate antioxidant capacity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
2.1. Chemical-based assays . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
2.1.1. Scavenging activity toward stable free radicals (DPPH• , ABTS• +) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
2.1.2. Reduction of metal ions (FRAP and CUPRAC assays) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
2.1.3. Competitive methods (ORAC and TRAP assays) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
2.1.4. Oxidation of low density lipoprotein (LDL) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
2.1.5. Nanoparticles-based assays . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
2.1.6. Other chemical assays . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
2.2. Evaluation of the antioxidant activity at cellular level . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
2.2.1. Cellular antioxidant activity (CAA) assay . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
2.2.2. Expression of antioxidant enzymes vs inhibition of pro-oxidant enzymes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
2.2.3. Activation vs repression of redox transcription factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
3. Correlation between chemical-based antioxidant capacity indexes and cellular-based assays . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
4. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9

Ana Denicola is Head of the Physical Biochemistry Camilo López Alarcón received his Doctorate in
laboratory at the Chemical Biology Institute of Fac- Chemistry in 2004 from the University of Chile, Chile.
ulty of Science, University of the Republic (UdelaR), He then gained a posdoctoral position at Dr. Eduardo
Uruguay. She graduated in Chemistry/Pharmaceutical Lissi‘s laboratory at University of Santiago of Chile,
Chemistry at the UdelaR, Uruguay, and received her where he focused on free radicals and antioxidants
PhD in Biochemistry at Virginia Tech, Va, USA. She is chemistry. At present, he is Associate Professor at
now Full Professor of Physical Biochemistry at the Fac- Pontificial Catholic University of Chile, where he is
ulty of Science, UdelaR. Her major research interests studying new methodologies to evaluate in vitro
concern mechanisms of bioproduction and reactivity antioxidant capacity of foods, beverages and human
of oxygen and nitrogen species, structural and func- fluids. His current research focuses on the interac-
tional characterization of oxidative modifications of tion of biologically-relevant compounds with reactive
proteins, synthetic and natural antioxidants. species such as nitrous acid, peroxyl radicals, super-
oxide, hypochlorite and nitrogen dioxide.

1. Introduction therefore, the obtained antioxidant capacity indexes not necessary


reflect the antioxidant effects that would be associated with a par-
During the last decades it has been proposed that oxidative ticular sample in vivo. The ability of natural products to induce
stress, defined as the unbalance between reactive oxygen and nitro- an antioxidant response at the cellular level also has to be eval-
gen species (ROS/RNS) production and the antioxidant defense, uated. A good antioxidant is not just a good radical scavenger and
plays a pivotal role in different pathophysiological conditions [1,2]. reducing compound but a molecule that can exert its antioxidant
Oxidative stress, originated from an increase in ROS/RNS produc- activity by activating transcription factors that induce the expres-
tion or from a decrease in the antioxidant network, is characterized sion of antioxidant enzymes, thus, ameliorating oxidative stress
by the inability of endogenous antioxidants to counteract the oxida- [15]. To evaluate a new product as antioxidant, as a compound able
tive damage on biological targets [3]. In this context, it has been to change the redox cellular state, we must use a cellular antiox-
suggested that an intake of a rich antioxidant diet is inversely idant assay since participation of different components of the cell
associated with the risk to develop some pathologies like car- are critical to finally develop an antioxidant response.
diovascular diseases [4–6]. Thus, attention has been paid on the In the present work we review the chemical-based method-
antioxidant capacity of natural products, with particular interest ologies employed for screening antioxidant capacity of natural
on those that are frequently (or potentially) consumed by peo- products discussing the classical and non-classical mechanisms
ple. Different in vitro chemical-based assays have been developed of action of antioxidant-containing natural products. In particu-
to determine the antioxidant capacity of natural products, includ- lar, we highlight the advantages and drawbacks of each method;
ing the popular ORAC, DPPH• scavenging method, ferric reducing introduce the importance of pursuing the evaluation of antioxidant
capacity, etc. and more recently the use of nanoprobes to evaluate capacity at the cellular level and compare reported results between
the metal-reducing capacity of antioxidants [7–14]. These assays chemical- and cellular-based assays.
are based on different strategies and endow different informa-
tion about the ROS/RNS-sample interaction. However, to study the
potential antioxidant health-protecting effects of natural products, 1.1. Mechanisms of antioxidant actions
considering the complexity involved in their in vivo mechanisms
of action, a single in vitro chemical method is not enough to eval- 1.1.1. The classic (traditional) antioxidant mechanism
uate and compare their antioxidant properties. In addition, these The classical definition of antioxidant as “any substance that
chemical assays do not consider relevant parameters involved in delays, prevents or removes oxidative damage to a target molecule”
biological environments such as lipophilicity and bioavailability; [2] is usually understood as the ability of these compounds to
C. López-Alarcón, A. Denicola / Analytica Chimica Acta 763 (2013) 1–10 3

neutralize free radicals, acting for example, as chain-breaking presence of metals generates hydroxyl radical (Fenton mechanism),
derivatives in agreement with Reaction (1). a strongly oxidant free radical.

AH + FR• → A• + FRH (1) A• + O2 → AOO• (3)


•−
A• + O2 → Aox + O2 (4)
where AH and FR• represent an antioxidant and a free radical,
respectively.
1.1.2. New mechanisms based on redox signaling
Thus, Reaction (1) is considered the base of the classical mecha-
In 1985 Helmut Sies introduced the concept of oxidative stress
nism of action of antioxidants and explains their ability to inhibit (or
as “a disturbance in the prooxidant-antioxidant balance in favor
delay) many damaging processes induced by FR• on lipids, proteins,
of the former” [3]. Experimental evidence supported the idea that
or DNA. However, Reaction (1) does not represent all the factors
oxidative stress contributes to the development of several patholo-
affecting the antioxidant activity of a compound or an antioxidant-
gies including cardiovascular disease, neurodegenerative diseases,
containing mixture. Among these factors, the most relevant are: the
cancer and also aging. ROS such as superoxide, singlet oxygen, H2 O2
reactivity of antioxidants toward FR• , the number of FR• molecules
and hydroxyl radical were mainly considered responsible for these
neutralized by each antioxidant molecule (stoichiometric factor, n),
damaging oxidative reactions. More recently, new radical species
the liposolubility of the antioxidant, and the presence of secondary
were identified, now centered on nitrogen, thus termed reactive
reactions.
nitrogen species RNS, derived from the biologically produced rad-
ical, nitric oxide (• NO). The concept of ROS/RNS as just biologically
1.1.1.1. Reactivity and stoichiometric factor. Taking into account damaging species has begun to change. Enzyme systems produce
Reaction (1) as a bimolecular process, the rate of the reaction, r, reactive species not only for chemical defense or detoxification, but
expressed as: also for cell signaling and biosynthetic reactions. The presence of
r = k1 [AH] [FR• ]ss (1) both, toxic and beneficial effects of ROS/RNS precludes a simple
definition of oxidative stress. At the same time, the intervention
depends on the kinetic rate constant k1 , the antioxidant concen- with antioxidant supplements did not show the results expected
tration [AH], and the steady state concentration of FR• ([FR• ]ss ). [19–21], in essence because most pathologies are multifactorial
Accordingly, from a kinetic point of view, a compound with a high and oxidative stress is just one of many contributing factors. A
k1 value would be a good antioxidant. Nonetheless, for antioxidants new concept of oxidative stress was emerging, not limited to free
expected to act in biological environments (for example human radical damage of the macromolecular machinery but to pertur-
blood plasma) the latter is relevant only if their reactions with FR• bation of cellular redox status. Based on new accumulating data
are faster or comparable with the rate of endogenous antioxidants on redox signaling pathways, antioxidant intervention trials and
(for example human serum albumin or uric acid) reacting with FR• . oxidative stress markers, Dean Jones in 2006 re-defined oxidative
In that case, the stoichiometry of Reaction (1), defined as the num- stress as “a disruption in redox signaling and control” [15]. It is
ber of FR• molecules that each antioxidant is able to neutralize (n), clear today that the mechanism of action of antioxidants is more
could also be a relevant factor in a food or beverage preservative. complex than just intercepting reactive free radicals [15,22]. Free
radicals can be deleterious to life depending on the type of radical
1.1.1.2. Liposolubility. The capacity of a particular antioxidant to produced and the level and site of production, but at the same time,
reach the place where FR• are being generated is also a critical low concentrations of these reactive species are essential to per-
aspect. In this context, antioxidants with high in vitro antioxidant form normal physiological functions like gene expression, cellular
capacity are not necessarily efficient neutralizers of FR• in compart- growth and defense against infection. The redox cellular network
mentalized systems. This is a pivotal point when trying to inhibit is finely regulated and its perturbation provokes oxidative stress.
lipid peroxidation processes in cell membranes, where antioxi- The idea behind antioxidant supplementation is to restore redox
dants should have an appropriate liposolubility to be incorporated cellular status. Some studies have indicated that antioxidants could
into the membrane and to react with FR• through chain-breaking also have deleterious effects on human health depending on dosage
reactions [16]. For instance, ␣-tocopherol is liposoluble and is and bioavailability [19,20]. It is therefore necessary to explore the
the most important biological antioxidant in membranes, but its mechanism of action of a potential antioxidant at the cellular level
solubility in water is very low. One particular case is ascorbic in order to extrapolate its therapeutic potential (Fig. 1).
acid, which in spite of its well-known hydrophilic character is
able to work additively with membrane-immersed ␣-tocopherol 1.1.2.1. Discrete and compartmentalized redox signaling pathways.
to prevent lipid peroxidation, by reducing the surface-exposed ␣- Evidence has accumulated showing that endogenous oxidants like
tocopheroxyl radical back to ␣-tocopherol [17]. H2 O2 can act as second messengers and trigger a cascade of intra-
cellular responses resulting in the expression of antioxidant and
detoxifying enzymes in order to control the cellular redox status
1.1.1.3. Secondary reactions. Other aspect to consider in the antiox-
[23]. Much research is still needed in this area but some redox
idant behavior of a particular sample is the ability of secondary free
signaling pathways have already been identified. Two transcription
radicals (A• ) generated in Reaction (1) to damage biological targets
factors associated with redox control are: Nrf-2 (nuclear factor E2-
(BTH in Reaction (2)).
related protein 2) and NF-␬B (nuclear factor kappa B) with defined
A• + BTH → AH + BT• (2) redox control compartimentalized in cytosol and nucleus.
Nrf-2 is a redox-sensitive transcription factor that is activated
It has been observed that secondary phenoxyl radicals can, in by an oxidative signal in the cytoplasm that causes its transloca-
some conditions, trigger oxidative modifications on proteins, DNA, tion to the nucleus where it binds to DNA ARE-regions (antioxidant
as well as on cell membrane lipids [18]. Furthermore, depending on response elements) inducing the expression of cytoprotective
the antioxidant compound and/or the place where A• is generated, enzymes like glutathione S-transferase GST, superoxide dismu-
it can react with O2 to form a secondary peroxyl radical or superox- tase SOD, heme oxigenase-1 HO-1, NADPH-quinone oxidase NQO
ide anion (Reaction (3) and Reaction (4), respectively). In the latter (ARE-regulated genes) [24]. In the cytoplasm, Nrf-2 is associated
case, dismutation of superoxide (usually catalyzed by superoxide with Keap1 (Kelch-like ECH associated protein) that facilitates its
dismutase, SOD) yields hydrogen peroxide (H2 O2 ), which in the ubiquitination and degradation, thus, keeping down the levels of
4 C. López-Alarcón, A. Denicola / Analytica Chimica Acta 763 (2013) 1–10

– The ability of antioxidants to protect a target molecule exposed


to a free radical source.
– The capacity of antioxidants to inhibit the oxidation of low-
density lipoprotein LDL.

2.1.1. Scavenging activity toward stable free radicals (DPPH• ,


ABTS• +)
DPPH• (2,2-Diphenyl-1-picrylhydrazyl) and ABTS (2,2 -
Azinobis-(3-ethylbenzothiazole-6-sulphonate) radical cation,
ABTS• +) are two stable and colored free radicals that have been
widely employed to determine antioxidant capacity [26,27]. DPPH•
is commercially available, whereas ABTS• + must be generated
from the oxidation of ABTS by oxidants such as K2 S2 O8 , MnO2
or peroxyl radicals [28]. DPPH• is soluble in organic solvents and
presents a typical absorption band at 517 nm (in ethanol); while
ABTS• + is soluble in aqueous as well as in alcoholic media and its
absorption at 734 nm is used. Usually, the decrease in absorption
Fig. 1. Different mechanisms of action of antioxidants: (1) inhibition of oxidant intensity in the presence of antioxidant-containing samples is
enzymes, that translate in a decrease in the ROS/RNS cellular production, (2) interac-
registered after a fixed incubation time (30 min or less), thus no
tion with redox signaling pathways that translate in a cellular antioxidant response
(less classical view of an antioxidant compound or mixture), (3) direct reaction with kinetic data are considered. In this context, Sánchez-Moreno et al.
ROS/RNS yielding a less toxic/reactive product (the most popular view of an antiox- [29] introduced the antiradical efficiency (AE) parameter which
idant, as a “free radical scavenger”). Enzymes producing ROS/RNS: NOS (nitric oxide includes kinetic factors to characterize the reaction between
synthase), NOx (NADPH oxidase), COX (cyclooxygenase), MPO (myeloperoxidase).
DPPH• and polyphenols.
Antioxidant enzymes: CAT (catalase), SOD (superoxide dismutase), Prx (peroxire-
doxin), GPx (glutathione peroxidase), Trx (thioredoxin), TR (thioredoxin reductase),
The shortcomings of these assays are the complexity of the
GR (glutathione reductase). mechanisms of reaction, the dependence of the index with the
experimental conditions, and the poor correlation between DPPH•
and ABTS• + chemical structures with free radicals produced in bio-
Nrf-2. Activation of Nrf-2 by ARE-inducers provokes dissociation of logical systems [30].
Nrf-2/Keap1 complex, less ubiquitination and the corresponding
accumulation of Nrf-2 that reach the nucleus leading to increase
transcription of genes under control of ARE. Not only Keap1 has 2.1.2. Reduction of metal ions (FRAP and CUPRAC assays)
critical cysteine residues that are modified under oxidative stress, These methods aim to evaluate the capacity of the sample to
but also Nrf-2 needs a reduced cysteine to bind to DNA. reduce ferric or cupric ions in aqueous media. The FRAP (ferric
Compounds that increase the expression of Nrf-2 and/or reducing antioxidant power) assay, originally developed to eval-
facilitate the dissociation of Nrf-2/Keap1 and translocate Nrf-2 uate the ability of human plasma as antioxidant, uses the reaction
to the nucleus, provoke an antioxidant response since ARE- of tripyridyl triazine-ferric complex (FeIII -TPTZ) with antioxidants
genes are induced, i.e. the expression of antioxidant enzymes [31], while the CUPRAC (Cupric ion-Reducing Antioxidant Capac-
increased. ity) method, developed by Apak et al. [32], determines the ability
The NF-B family consists of a group of inducible transcrip- of a sample to reduce the neocuproine-cupric complex (CuII –Nc).
tion factors which regulate immune and inflammatory responses The basis of both methodologies is that the complexes of TPTZ or
and protect cells from undergoing apoptosis in response to cel- Nc with the reduced form of the metals present characteristic visi-
lular stress (including oxidative stress). NF-B is kept inactive ble absorption bands with maximal intensity at 593 and 450 nm for
in the cytoplasm by association with inhibitors I␬B proteins. In FRAP and CUPRAC assay, respectively. Thus, the capacity of a sam-
response to inflammatory stimulus, such as oxidants, I␬B proteins ple to reduce the metal complexes generating the corresponding
are rapidly degraded by the proteasome liberating NF-␬B protein visible absorption bands in a fixed incubation time, is employed to
to the nucleus where it binds to specific DNA sequences, activat- determine FRAP or CUPRAC values. FRAP assay requires an acidic
ing the expression of specific pro-inflammatory and anti-apoptotic pH (3.6), that is far from the physiological pH. In contrast, CUPRAC
genes [25]. assay, performed at pH 7.0, better simulate physiological condi-
Compounds that decrease the expression of NF-␬B and/or tions. In addition, CUPRAC assay has shown to differentiate the
inhibit its activation prevent its translocation to the nucleus and reducing power of thiol-type antioxidants [33].
the induction of pro-inflammatory/pro-oxidant genes. The main advantage of these methods is the simplicity of the
experimental conditions. However, as DPPH• and ABTS• + -based
2. Methodologies to evaluate antioxidant capacity assays, FRAP and CUPRAC indexes depend on the reaction time.
Depending on the sample under study, different endpoints of the
2.1. Chemical-based assays reaction can be registered. In this context, it has recently been pro-
posed a kinetic matching approach to express antioxidant capacity
Taking into account the complexity involved in the in vivo action in a more standardized way [34].
of antioxidants, different in vitro methodologies have been devel-
oped to estimate, in a simple experimental way, the capacity of
2.1.3. Competitive methods (ORAC and TRAP assays)
antioxidants and their complex mixtures to interact with (neutral-
These methodologies evaluate the ability of a particular sam-
ize) ROS/RNS. The assays are based on diverse strategies aimed to
ple to inhibit the consumption of a target molecule (usually
evaluate:
followed by UV–visible absorption or fluorescence spectroscopy)
mediated by peroxyl radicals. Usually, AAPH (2,2 -Azobis (2-
– The consumption of stable free radicals by antioxidants. methylpropionamidine) hydrochloride, also abbreviated ABAP) is
– The capacity of antioxidants to reduce cupric or ferric ions. employed as peroxyl radical (ROO• ) source, that generates ROO•
C. López-Alarcón, A. Denicola / Analytica Chimica Acta 763 (2013) 1–10 5

2.1.3.2. TRAP (total radical-trapping antioxidant parameter) assay.


This method is based on the oxygen uptake associated to the per-
oxidation process of human plasma [43]. In particular, the method
evaluates the induction period (lag time ), generated in the kinetic
profiles of oxygen uptake during the incubation of human plasma
with AAPH (Eq. (2)).

TRAP = Ri xplasma (2)

where Ri is the rate of peroxyl radicals formation and  plasma is


the lag time in oxygen consumption generated by the presence of
human plasma.
Thus, taking into account Eq. (2), TRAP index is defined as
the number of moles of ROO• trapped per liter of fluid (plasma)
[43,44]. Different variations have been proposed to assess the TRAP
index of natural products. For example, the use of luminol (o-
aminophthaloylhydrazide) or pyranine (8-hydroxy-1,3,6-pyrene
Fig. 2. Protection of fluorescein oxidation (induced by ROO• ) elicited by Peumus-
trisulfonic acid) as probes to determine this index. The reaction
boldus extract. Control experiment (in the absence of extract, ), Peumusboldus
0.1 ␮L/mL (); 0.3 ␮L/mL (); 0.5 ␮L/mL (♦). Data taken from [40]. of luminol with ROO• emits photons that can be measured in a
luminometer, while pyranine oxidation by ROO• can be followed
by fluorescence [44,45]. It is important to consider that TRAP
at a known rate in the first hours of incubation in aqueous media, index reflects mainly the number of ROO• trapped per antioxidant
according to Reaction (5) [35] molecule, thus, this is an index exclusively related to the stoichiom-
etry of the ROO• -antioxidant reaction.
AAPH + O2 → 2ROO• + N2 (5)

The incubation of AAPH with a particular target molecule (TM) 2.1.4. Oxidation of low density lipoprotein (LDL)
leads to a change in the UV–visible absorption or fluorescence The oxidation of LDL mediated by ROS/RNS has been studied
intensity of that TM. Thus, the co-incubation of TM, AAPH and pure since many years ago. At present, it is well known that ROS play a
antioxidants or their complex mixtures (XH) usually leads to the pivotal role in the initiation, propagation and termination reactions
inhibition of TM consumption. The minimal reactions involved in of the LDL lipid peroxidation processes [2]. In vitro assays usually
these assays are: employ cupric sulfate as initiator of LDL oxidation and the lipid per-
oxidation processes are easily followed by UV spectroscopy and/or
ROO• + TM → consumption (6)
chemiluminiscence techniques [46]. In the first case, the formation
ROO• + XH → X• + ROOH (7) of diene conjugates at 234 nm is followed, while in the second one,
the emission of photons related to the formation of final oxida-
2ROO• → non radicals products (8) tive products is measured. When cupric sulfate is added to a LDL
solution, the kinetic profiles are characterized by the presence of a
2.1.3.1. ORAC (oxygen radical absorbance capacity) assay. Among lag time associated with the presence of endogenous antioxidants
the methodologies developed to estimate antioxidant capacity, (mainly vitamin E and coenzyme Q) in the LDL particle. After that
this assay is one of the most employed. In fact, this method has period, the peroxidation of lipids is evidenced as an increase in
been used for estimating the antioxidant capacity of the mainly the absorbance at 234 nm that eventually reaches a plateau, after
consumed foods and beverages in USA [36]. ORAC assay is an minutes or hours. In the presence of antioxidants, this lag time is
integrative method based on Reactions (5)–(8). In this assay the increased, implying an additional protection of LDL given by the
antioxidant capacity is evaluated from the area under the curve sample. Thus, measurement of the lag time has been frequently
(AUC) of the kinetic profiles of the TM consumption (Fig. 2). used to evaluate antioxidant capacity.
AUC values are commonly compared with gallic acid or Trolox (a The main advantage of this in vitro assay is the use of a biologi-
hydrosoluble vitamin E analog) allowing determine an ORAC index cal relevant target. One of the most important shortcomings of this
in terms of these reference compounds. method is the variability between lot to lot of the LDL sample. This is
The ORAC assay, proposed by Cao et al. in 1993 was originally a consequence of the complexity of the LDL particle, which includes
applied employing beta-phycoerythrin as TM [37]. Nonetheless, proteins (apoB), lipids (phospholipids, saturated and unsaturated
in 2001 Prior’s group proposed an improved ORAC assay employ- lipids, triglycerides, free and esterified cholesterol), and endoge-
ing fluorescein as probe which is photostable, cheap, and does not nous antioxidants (vitamin E, coenzyme-Q), that can vary from
interact with polyphenols [38]. donor to donor. Also, when cupric sulfate is employed as initia-
Considering Reactions (5)–(8), the ORAC index is expected to tor of oxidation, it should be considered that the antioxidant effect
be independent from the TM employed. However, reports employ- can also derive from the copper-chelating capacity of the sample.
ing pyrogallol red and other compounds as TM, have shown that
the selection of the TM can influence significantly this index [39]. 2.1.5. Nanoparticles-based assays
Furthermore, in some cases, depending on the selected TM, contra- More recently, novel chemical assays based on nanotechnology,
dictory results were obtained [40]. in particular the use of nanoparticles, have been proposed to evalu-
The ORAC assay has the advantage to be a simple and stan- ate antioxidant capacity. In pioneering work, Scampicchio et al. [13]
dardized method, however, secondary reactions or even reactions estimated the antioxidant power of several phenolic acids from the
associated with repair mechanisms can be present [41]. In addi- generation and growth of gold nanoparticles (AuNPs) from a AuIII
tion, it has recently been reported a re-examination of the ORAC solution (HAuCl4 ) by the appearance of a sharp plasmon absorp-
assay that indicates that antioxidant-metal reactions could result tion band at 555 nm. The optical properties of the generated AuNPs
in a lower concentration of antioxidants and therefore to an under- correlated well with the reduction potential of the phenolic acids
estimation of the ORAC value [42]. as measured by cyclic voltametry, and it was proposed as a form
6 C. López-Alarcón, A. Denicola / Analytica Chimica Acta 763 (2013) 1–10

to evaluate the antioxidant capacity of pure compounds and com- of the antioxidant capacity index [48]. For instance, polyphenols
plex mixtures. For example, this experimental approach has been have been observed to inhibit xanthine oxidase activity using a
recently employed by Liu et al. [47] to evaluate the antioxidant parallel CUPRAC assay [49].
capacity of chrysanthemum extracts and tea beverages. In addi- Hydroxyl radical (• OH) is one of the most reactive free radicals
tion, Vilela and collaborators [14], following the generation of AuNP and the strongest oxidant, thus in vivo, its toxicity depends strongly
at 540 nm (A540 ) described a sigmoidal behavior as a function of on its site of formation. Its high reactivity results in a very short
polyphenol concentration (X) that fit the equation: diffusion range that may prevent reaction with a critical biological
target. In a general context, the proposed methods employ a • OH-
A max
A540 =   (3) sensitive molecule such as 2-deoxy-D-ribose, fluorescein, or DMPO
−KAuNPs X−X 50
1+e C and a source of • OH, usually a system containing ferric sulfate-
H2 O2 -ascorbic acid or Fenton-like reactions [7,9]. In the case of
where Xc50 represents the concentration of polyphenol that gives fluorescein, the consumption of this probe in the absence and pres-
half maximum plasmon resonance absorption and KAuNPs the ence of the sample under study is estimated by fluorescence, while
number of AuNPs produced per polyphenol concentration unit. in the case of deoxyribose as a probe, the formation of colored
The authors proposed to use KAuNPs as a parameter to estimate adducts with thiobarbituric acid are determined. In a similar way
antioxidant capacity. The assay has been applied to evaluate the to superoxide, the inhibition of the DMPO-OH adduct formation by
antioxidant activity of natural products such as tea, apples, pears, ESR has also been used. It is worth to point that, considering the
red wines and honey. Interestingly, a good correlation between extraordinary high reactivity of • OH toward biological targets and
KAuNPs and total phenolic content has been observed [14]. many organic compounds, it is somehow irrelevant to evaluate the
In addition to the latter studies, Özyürek et al. [12] have reported capacity of a sample as • OH scavenger in vitro. Only if the compound
a novel methodology employing silver nanoparticles to assess is present at a high concentration in the site of cellular formation
antioxidant capacity. The method, named SNPAC (Silver NanoParti- of • OH, will it efficiently compete for • OH.
cle Antioxidant Capacity) employs Trolox as standard antioxidant, Peroxynitrite (ONOO− ), the product of the diffusional reac-
and reflects the total antioxidant capacity (TAC) of the sample under tion between superoxide anion and nitric oxide, is implicated in
study. The assay is based on the ability of polyphenols to reduce different pathological conditions [50]. As in the other cases, the
Ag+ ions in the presence of citrate-stabilized silver seeds, and eval- reaction of antioxidants with ONOO− (or the free radicals derived
uates the intensity of the plasmon visible-absorbance at 423 nm to from homolysis of peroxynitrous acid, ONOOH) has been assessed
quantitatively determine antioxidant capacity. In comparison with employing probe molecules. Dihydrorhodamine, dihydrofluores-
previous reports the novelty of this assay is to employ a first step cein, pyrogallol red and tyrosine have been used. Oxidation of
reduction of Ag+ ions by citrate to form silver seeds. Afterwards, the the first two probes is followed by fluorescence increase [51],
addition of the polyphenol-containing sample increase the plas- while pyrogallol red oxidation is monitored by decrease in absorp-
mon absorption intensity. The role of polyphenols as secondary tion at 540 nm. 3-Nitrotyrosine formation as a footprint of ONOO-
reducing agents would imply a more robust and reproducible (although other • NO2 -dependent biological systems can also per-
methodology than the assays employing a direct reduction of form this modification) can be followed by HPLC-MS, UV–vis
metal ions by antioxidants. Interestingly, the authors describe that spectroscopy, or even immunochemistry [52,53]. It is worth to
growth but not nucleation of silver nanoparticles showed a linear point that the biological nitration of tyrosine residues is a radical
concentration-dependent response. The SNPAC method has been process initiated by NO-derived species and not by oxygen radicals
applied to a wide variety of pure antioxidants and complex mix- [53] that it was not considered (nor quantified) in the classical free
tures such as fruit juices and herbal teas. As advantages, the method radical scavenging methods.
has shown good linearity with polyphenol concentration and has Hypochlorite ion (ClO− ) and its protonated form, hypochlor-
not been affected by the presence of reducing sugars, fruits acids, ous acid (HOCl), are oxidative species generated by the enzyme
nor amino acids present in the extracts. myeloperoxidase in activated neutrophils. Hypochlorite is a
These new nanoparticles-based assays to determine antioxidant powerful oxidizing agent able to damage biomolecules and/or
capacity of natural products comprise a novel and promising area cellular structures during processes associated with inflamma-
combining nanoscience with food and health research. tion or degenerative diseases [2]. Different approaches have been
employed to evaluate the scavenging activity of antioxidants or
2.1.6. Other chemical assays their complex samples toward hypochlorite, being the use of
In addition to the above discussed assays, other methodologies proteins or low-molecular weight targets the strategy most fre-
have also been proposed to evaluate antioxidant capacity. These quently employed. For instance, ␣1-antiproteinase, myoglobin
methods aim to evaluate scavenging activity toward ROS/RNS and serum albumin have been used as protein targets, while
formed in vivo like superoxide anion, hydroxyl radical, peroxyni- 5-thio-2-nitrobenzoic acid (TNB), 5-aminosalycilic acid, fluores-
trite and hypochlorite. cein and luminol have been employed as hypochlorite-oxidizable
The classical approach to determine antioxidant capacity low-molecular weight target molecules [9]. The inhibition given
toward superoxide employs the xanthine-xanthine oxidase sys- by antioxidants, on the consumption of such targets, is followed
tem as the superoxide source and probes such as ferricytochrome evaluating chloramines formation, by changes in both UV–visible
c or nitrobluetetrazolium (NBT) [2,7,9]. The reduction of both absorption and/or fluorescence as well as chemiluminescence.
probes, mediated by superoxide, is followed by visible absorbance,
assessing the formation of ferrocytochrome c or formazan, respec- 2.2. Evaluation of the antioxidant activity at cellular level
tively. Alternatively or in conjunction, the reactivity toward
superoxide is determined by electron spin resonance (ESR) employ- Going beyond the in vitro chemical-based assays for testing
ing 5,5-dimethyl-1-pyrroline-N-oxide (DMPO) as spin-trap [2,7]. potential antioxidants is more expensive because it requires com-
In the presence of an antioxidant-containing sample, the reduction plex cellular testing systems or full clinical trials analyzing blood
of ferricytochrome c, NBT or the formation of DMPO-OH adduct samples before and after consuming the product. Nevertheless,
is inhibited. These assays have been widely employed, however, it is very important to proceed to cellular assays after screening
it is important to consider that some antioxidants, particularly antioxidant activity with an in vitro chemical method in order
flavonoids, can inhibit xanthine oxidase leading to overestimation to approach some aspects of the bioavailability of the potential
C. López-Alarcón, A. Denicola / Analytica Chimica Acta 763 (2013) 1–10 7

different ways, either directly reacting with ROO• or secondary


oxidants ROS/RNS, or indirectly, inducing an antioxidant cellular
response that in sum reduce the levels of oxidant cellular status.
The CAA assay protocol is detailed in [54]. Briefly, HepG2 cells are
seeded on a microplate at a density of 6 × 104 cell/100 ␮L/well. After
24 h of growth, cells are incubated with the natural product extract
and the probe dichloro-dihydrofluorescein diacetate (DCFH2 -DA,
25 ␮M) for 1 h at 37 ◦ C. The ester is used so that it permeates
the cellular membrane and once inside the cell is hydrolyzed by
endogenous esterases to the more polar DCFH2 . Then, 600 ␮M
AAPH is added to each well and fluorescence is recorded every 5 min
for 1 h (exc = 485 nm, em = 535 nm). CAA values are calculated as:

 
SA
CAA = 1 −  (4)
CA

where ʃSA is the integrated area under the curve sample fluores-
cence vs time and ʃCA is the integrated area from the control curve.
Quercetin is used as standard and CAA values are expressed as
Fig. 3. Schematic representation of the basis of the cellular antioxidant activity
␮mol equivalents of quercetin per 100 ␮mol of compound (pure
(CAA) assay. Cells are loaded with the redox-sensitive probe DCFH2 (the ester form
DCFH2 -DA is supplied so that to permeate the membrane and once inside the cell is flavonoid) or 100 g product (fruit, vegetable) or per 100 ␮mol of
hydrolyzed by endogenous esterases). Cells are incubated with the natural product total phenolics (considering all phenol content as expressed as gal-
extract or just medium (control) and exposed to AAPH that generates a flux of per- lic acid equivalents).
oxyl radicals (ROO• ). The antioxidant compounds can protect DCFH2 from oxidation
Among the pure compounds examined in the CAA assay,
(thus diminish fluorescence) by different mechanisms: (1) scavenge peroxyl radi-
cals in the membrane diminishing lipoperoxidation, (2) react with AAPH avoiding quercetin showed the highest CAA activity, followed by kaempferol,
intracellular ROO• formation, (3) compete with DCFH2 for oxidants ROS/RNS, (4) myricetin, epigallocatechin gallate and luteolin. In contrast, caffeic
react with ROO• preventing other radicals formation, (5) inhibit a redox pathway acid, gallic acid and ascorbic acid showed less than 1% CAA activity
toward formation of ROS/RNS that oxidize DCFH2 . [54].
Cellular uptake of DCFH2 -DA is rapid and relatively stable
antioxidant like uptake, metabolism, partitioning in membranes, although leakage is observed after 1 h, thus long runs should be
that are crucial to the effectiveness of the antioxidant in vivo. avoided [55]. Exposure of loaded cells to light should be minimized
Animal models and human studies are more appropriate but more to avoid artifactual generation of superoxide and oxidation of the
expensive and time-consuming, making the cell culture assays probe [56]. The use of dark microplates is recommended. We should
very attractive as intermediate testing methods. not forget that working with cells is not like isolated chemical reac-
According to the new definition of an antioxidant, a redox-active tions. The cell response is quite complex, thus, more variable results
compound or mixture able to modulate the redox status of the cell, are expected depending on the assay conditions like growth status
it is critical to use cellular assays in order to evaluate the potential of the cell line, endogenous antioxidant levels and initial ROS/RNS
antioxidant activity of a compound or extract. Antioxidant action production. Several replicas should be performed in order to mini-
is not limited to ROS/RNS scavenging but includes upregulation mize these variables.
of antioxidant and detoxifying enzymes, modulation of redox cell Since the publication of the CAA assay by Wolfe and Liu in
signaling and gene expression (Fig. 1). 2007 [54], this method has been applied to several natural prod-
uct extracts, food and dietary supplements. Among fruits analyzed,
2.2.1. Cellular antioxidant activity (CAA) assay the ones with higher phenolics content were the ones display-
Wolfe and Liu developed a cellular antioxidant activity (CAA) ing higher CAA values; berries (blueberry, cranberry, etc.) with
assay to evaluate the antioxidant activity of food extracts and >50 ␮mol quercetin/100 g followed by apples, and grapes with less
dietary supplements [54]. They use human hepatocarcinoma than 10 ␮mol quercetin/100 g [57].
HepG2 cells loaded with the redox sensor dihydrodichlorofluores- Different cell types have been used for the CAA assay besides
cein DCFH2 that easily oxides to fluorescent dichlorofluorescein HepG2, including Caco-2 matured differentiated intestinal cells
DCF by ROO• (from AAPH decomposition). The method measures [58], human gastric adenocarcinoma cell line AGS with rapid pro-
the ability of compounds to prevent the oxidation of intracellu- liferation properties [59], vascular endothelial cells EA.hy926 [60],
lar DCFH2 that can be followed by fluorescence (exc = 485 nm, human macrophage cell line U937 [61], human lung fibroblasts
em = 535 nm) and the results are expressed in ␮moles of quercetin (WI38, IMR-90) [62]. An interesting one is erythrocytes, since it
equivalents. The CAA assay is an important tool for screening is easily available and testing the potential protection of red blood
antioxidant activity in natural products extracts that evaluates its cells (RBC) is biologically relevant per se as RBC play a critical role
potential to exert an antioxidant response at the cellular level, in reducing oxidative stress in the vasculature [63,64]. The same
not just its capacity as a reducing agent. The principle of the principle is used, i.e., cells are loaded with the redox probe DCFH2 ,
method is depicted in Fig. 3. ROO• generated from temperature- incubated with the antioxidant to be tested and exposed to oxida-
decomposition of the azo compound AAPH can be produced at the tive stress via generation of ROO• with AAPH. Cellular oxidative
cell membrane or intracellularly and either directly oxidize DCFH2 stress can also be generated by cell culture exposure to H2 O2 in the
or produce other oxidants (ROS/RNS) that finally oxidize the probe. mM range (although plasma membrane is permeable to H2 O2 , there
It is worth to mention that DCFH2 is oxidized by ROO• but also other is a significant drop on H2 O2 concentration achieved intracellu-
biologically-produced ROS/RNS are capable to perform this oxida- larly). Again, the oxidation of the probe is followed by fluorescence
tion, like H2 O2 in the presence of peroxidases (not H2 O2 alone), [65,66]. RBC do not have nucleus nor mitochondria, thus the antiox-
peroxynitrite or hydroxyl radical [51]. The antioxidant compound idant cellular response in this type of cells will not involve gene
can prevent DCHF2 oxidation, thus reduce fluorescence levels in expression regulation but will consider the membrane permeation
8 C. López-Alarcón, A. Denicola / Analytica Chimica Acta 763 (2013) 1–10

of the compounds as well as interaction with cytosolic antioxidant of • NO from iNOS should be avoided to diminish the inflamma-
systems. tory state, while active production from eNOS at the endothelium
The properties of the cell line used in the CAA assay could is desirable to promote a healthy vascular function.
affect the result. The use of transformed cell lines has been criti- The beneficial effect of some polyphenols to increase eNOS
cized because they have altered antioxidant enzymes expression activity and at the same time inhibit endothelial Nox, translates
(for example increased catalase mRNA expression in HepG2) and into an effective increase of • NO bioavailability, since not only
performed asymmetrical cell divisions [65]. In addition, some cell more • NO is been produced by eNOS but also the formation of
lines (like MCF-7) contain estrogen receptors that could be acti- peroxynitrite from the reaction of • NO with superoxide is being
vated by natural products with phytoestrogen activity. prevented [78,81,82]. Increased • NO bioavailability at the endothe-
Another cellular-based assay uses Saccharomyces cerevisiae lium is likely responsible for the beneficial effect observed at the
as a simple model to evaluate antioxidant capacity of natural cardiovascular level by a diet rich in polyphenols [5,83].
products in living systems [67–69]. Exponentially growing yeast Enzymes involved in the metabolism of glutathione (GSH) are
(OD600 = 0.5–0.7) is exposed to H2 O2 to induce oxidative stress, important to maintain cellular redox status. In particular, glu-
in the absence and presence of serial dilutions of the extract tathione reductase (GR) regulates the balance between reduced
(non-toxic final concentrations) and cell viability is determined by (GSH) and oxidized (GSSG) glutathione at the expense of NADPH.
plating in solid YPD medium and counting the colonies. S. cerevisiae The GSH/GSSG ratio is used as an index of oxidative stress. Acti-
is sensitive to H2 O2 insult (only 45% survived after 1 h incubation vation of GR raises the level of GSH, thus reduces oxidative stress
at 28 ◦ C/160 rpm with 0.75 mM H2 O2 ). Pretreatment with isolated and in addition recycles GPx, glutathione peroxidase, an important
flavonoids [67–69] or complex polyphenolic mixtures like wine H2 O2 -detoxifying enzyme. Baroni et al. observed that GR and GPx
[70,71] partially suppressed the damage triggered by H2 O2 . activities were increased in yeast cells when exposed to wine in
the presence of H2 O2 that could explain the observed increased
2.2.2. Expression of antioxidant enzymes vs inhibition of survival rates [70].
pro-oxidant enzymes The thioredoxin system (thioredoxin Trx, thioredoxin reductase
The previous CAA assay evaluates the capacity of individual TR and peroxiredoxins Prx) is tightly connected to the GSH sys-
compounds or complex mixtures to effectively reduce the intracel- tem and also contributes to maintain the redox status of the cell.
lular oxidative state (the levels of the internal biomarker oxidized Increased Trx activity have been found in S. cerevisiae treated with
fluorescein are lower) but it does not say about the mechanism the green tea extract due to activation of Yap1 transcription factor by
antioxidant compound(s) used to reduce the oxidative stress: is it catechins present in the extract [84].
reacting directly with a radical oxidant?, which one?, inhibiting the It is interesting to point that among polyphenols, curcumin and
production of more radical oxidants?, repairing the biomolecules some flavonoids such as myricetin, quercetin and catechin, have
oxidized by a radical oxidant?. Nevertheless, it is an index that bet- been identified as potential anticancer agents with a mechanism
ter reflects the new concept of an antioxidant: a compound (or of action that involves strong specific inhibition of TR activity in
mixture) that modulates the cellular redox state. But we can go cancer cells and it is currently a focus of attention [85–87].
further and investigate if the compound displaying a good CAA
is in fact inhibiting an oxidase (for example NADPH oxidase that
2.2.3. Activation vs repression of redox transcription factors
produces superoxide) or inducing the expression of an antioxidant
If the antioxidant is able to interact with a redox transcription
enzyme (for example SOD), resulting in both cases in the reduction
factor, up-regulating the expression of antioxidant enzymes, it is
of superoxide/H2 O2 levels.
effectively reducing the oxidative status of the cell, and the inter-
NADPH oxidases are heme-flavoenzymes that produce ROS [72].
esting point of this mechanism is that the antioxidant response
The classical Nox (gp91phox ) also termed Nox2, is found in the mem-
is amplified. We need just a few molecules of the transcription
brane of phagocytes and forms an active complex with the other
factor modified in order to express an antioxidant enzyme that
membrane subunit p22phox as well as soluble regulatory proteins,
will catalytically reduce the reactive oxidant. Instead, the amount
to catalyze the production of superoxide anion that plays a crucial
of antioxidant compound needed to obtain the same response by
role in host defense. We now know that there is an entire family
directly reducing the radical oxidant is much higher, even with
of NADPH oxidases (Nox) that mediate diverse functions including
a strong reducing compound. Nevertheless, the free radical scav-
cell growth, apoptosis, innate immunity, angiogenesis, regulation
enging capacity of an antioxidant is a rapid pathway to protect
of the extracellular matrix and thyroid hormone biosynthesis. Nox
biological targets from oxidation while the effect mediated by up-
1–5 produce superoxide from molecular oxygen at the expense
regulating antioxidant enzymes is much slower, depending on the
of NAD(P)H, whereas Duox enzymes are known to release H2 O2
biosynthesis of new proteins.
without forming detectable amounts of superoxide.
Isolated polyphenols (like curcumin and quercetin) as well as
Certain polyphenols and metabolites are able to inhibit Nox
natural products extracts have been found to exert an antioxidant
activity reducing the endogenous production of ROS [73,74]. In
response via activation of Nrf-2 [88–91].
addition, complex mixtures from plant extracts or natural prod-
In addition, inhibition of Nf-kB activation rendering an
ucts like wine, cocoa, propolis, have been demonstrated to inhibit
anti-inflammatory/anti-oxidant response has been obtained by
this enzyme, in particular the Nox1 and/or Nox 4 isoforms, respon-
incubation of cell cultures to phenolic compounds like curcumin
sible for the constant production of superoxide at the endothelium
[92] or fruit extracts like blueberries [93]. Even more, amelioration
[75–79].
of inflammatory processes were observed in mice supplemented
Further examples of inhibition of key pro-oxidant enzymes by
with a diet rich in blueberries extracts associated with inhibition
polyphenols have been given, e.g., with 5-lipoxygenase and xan-
of Nf-kB activation [93].
thine oxidase [49,77,79].
Nitric oxide synthases (NOS) are a family of heme-flavoenzymes
that produce the radical • NO from l-arginine using NADPH and 3. Correlation between chemical-based antioxidant
molecular oxygen (also requires tetrahydrobiopterine BH4 and capacity indexes and cellular-based assays
Ca2+ /calmodulin) [80]. Different isoforms have been identified:
NOS1 (neuronal nNOS), NOS2 (inducible iNOS present in phago- As mentioned above, Liu’s group proposed an interesting assay
cytes) and NOS3 (endothelial eNOS). A prolonged high production to evaluate antioxidant capacity in cell cultures [54,57,94]. This
C. López-Alarcón, A. Denicola / Analytica Chimica Acta 763 (2013) 1–10 9

Considering that chemical and cell-based assays are influenced


by very different factors, it is not surprising that the antioxidant
capacity of a sample determined by cell-based assays does not
correlate well with their chemical indexes. Moreover, the results
obtained from chemical-based assays depend on the method
employed since they explore different chemical aspects of the
potential antioxidant compound(s). For example, a particular sam-
ple could yield a high FRAP index because it contains a good metal
reducing compound but it could be a poor scavenger of peroxyl
radicals, thus showing a low ORAC value. In addition, different com-
ponents of the natural product extract may have synergistic effects
and the antioxidant response will depend on the particular phe-
nolic profile. As an example, in Fig. 4 we analyze data previously
reported by Wolfe et al. [94]. As is depicted in Fig. 4A, ORAC values
of pure flavonoids do not correlate well with their ability to inhibit
the peroxyl radical-mediated damage on HepG2 cells (CAA index).
Some structures displaying high ORAC values did not show the cor-
responding high CAA indexes. Similarly, some compounds with low
ORAC index showed surprisingly high response in the CAA assay.
In addition, CAA values reported so far for isolated flavonoids, did
not correlate well with their partition coefficients (Fig. 4B), suggest-
ing that membrane/water partition of the antioxidant compound is
not the only factor affecting the antioxidant capacity in cell-based
systems.

4. Conclusions

Considering the diversity of mechanisms an antioxidant com-


pound or mixture can exert in vivo, it is not possible to find a single
analytical method to evaluate its antioxidant capacity. It is nec-
essary to apply more than one in vitro chemical-based assay that
evaluates different aspects of the reactivity of the compound(s)
toward ROS/RNS. It is very important to standardize the analytical
methods used (indexes change depending on the assay conditions)
and always express the results as equivalents of a standard in order
to make possible the comparison between samples. It is essential
Fig. 4. (A) CAA values vs ORAC indexes for isolated flavonoids (quercetin,
kaempferol, myricetin, morin, luteolin, epicathechin, catechin, galangin). (B) CAA
to add a cellular-based assay to evaluate the potential of the sam-
values vs logP defined as the octanol-water partition coefficient for the selected ple to elicit a cellular antioxidant response besides its potential
flavonoids. Data taken from [94]. (䊉) represents CAA values using a protocol that as a good scavenger of ROS/RNS. For testing natural products as
includes washing of the cells before peroxyl radical exposure. () represents CAA antioxidants, a sequential multifaceted approach is highly recom-
data without previous wash. See [94] for experimental details.
mended, including for example determination of total polyphenolic
content with Folin reagent, a competitive chemical assay like ORAC,
metal-reducing activity like FRAP or SNPAC, inhibition of LDL per-
method, named cellular antioxidant activity (CAA) assay, was born
oxidation, inhibition of tyrosine nitration, and importantly, a CAA
from the need for developing standard methods that allowed
assay. The antioxidant capacity of a natural product extract will
establish the antioxidant capacity of samples in biologically more
essentially depend on the bioavailability of the specific mixture of
relevant conditions. CAA index reflects the capacity of antioxidants
compounds present, their synergistic interactions to yield the final
to reduce intracellular oxidative stress and evaluates more than
antioxidant response at the cellular level.
the reduction potential of an antioxidant compound and its free
radical scavenging ability but also takes into account other fac-
tors like membrane permeability, cell uptake, distribution and Acknowledgements
metabolism. Therefore, unlike the classical antioxidant capacity
assays which mainly focus this issue from a chemical point of view, The authors thank Dr. Matías N. Möller for critical reading of the
CAA methodology has a more biological approach, in agreement manuscript and assistance on Figures. The work was supported by
with the new concept of antioxidant, as a compound able to mod- CSIC, UdelaR, Uruguay and FONDECYT (grant no. 1100659), Chile.
ulate the redox state of the cell.
This does not mean that chemical methods should be considered
References
useless when compared to cell-based assays. On the contrary, they
should be understood as complementary methodologies and it is [1] S.J. Flora, Cell Mol. Biol. 53 (2007) 1–2.
highly recommended to analyze the antioxidant capacity of natural [2] B. Halliwell, J.M.C. Gutteridge, Free Radicals in Biology and Medicine, 4th ed.,
Oxford University Press, New York, 2007.
products by using more than one chemical-based as well as cellular-
[3] H. Sies, Oxidative Stress: Introductory Remarks, Academic Press, London, 1985.
based assay. In this context, Honzel et al. [65] proposed a sequential [4] H.C. Hung, K.J. Joshipura, R. Jiang, F.B. Hu, D. Hunter, S.A. Smith-Warner, G.A.
strategy to determine antioxidant capacity, including ORAC assay, Colditz, B. Rosner, D. Spiegelman, W.C. Willett, J. Natl. Cancer Inst. 96 (2004)
evaluation of the protection given by the sample to erythrocytes 1577–1584.
[5] K.J. Joshipura, F.B. Hu, J.E. Manson, M.J. Stampfer, E.B. Rimm, F.E. Speizer, G.
(CAP-e method) and response of polymorphonuclear cells (PMN) Colditz, A. Ascherio, B. Rosner, D. Spiegelman, W.C. Willett, Ann. Intern. Med.
exposed to ROS. 134 (2001) 1106–1114.
10 C. López-Alarcón, A. Denicola / Analytica Chimica Acta 763 (2013) 1–10

[6] L.M. Oude Griep, J.M. Geleijnse, D. Kromhout, M.C. Ocke, W.M. Verschuren, PLoS [53] R. Radi, Proc. Natl. Acad. Sci. U. S. A. 101 (2004) 4003–4008.
One 5 (2010) e13609. [54] K.L. Wolfe, R.H. Liu, J. Agric. Food Chem. 55 (2007) 8896–8907.
[7] D. Huang, B. Ou, R.L. Prior, J. Agric. Food Chem. 53 (2005) 1841–1856. [55] J.A. Royall, H. Ischiropoulos, Arch. Biochem. Biophys. 302 (1993) 348–355.
[8] Z.Q. Liu, Chem. Rev. 110 (2010) 5675–5691. [56] E. Marchesi, C. Rota, Y.C. Fann, C.F. Chignell, R.P. Mason, Free Radic. Biol. Med.
[9] L.M. Magalhaes, M.A. Segundo, S. Reis, J.L. Lima, Anal. Chim. Acta 613 (2008) 26 (1999) 148–161.
1–19. [57] K.L. Wolfe, X. Kang, X. He, M. Dong, Q. Zhang, R.H. Liu, J. Agric. Food Chem. 56
[10] E. Niki, Free Radic. Biol. Med. 49 (2010) 503–515. (2008) 8418–8426.
[11] R.L. Prior, X. Wu, K. Schaich, J. Agric. Food Chem. 53 (2005) 4290–4302. [58] M. Sessa, R. Tsao, R. Liu, G. Ferrari, F. Donsi, J. Agric. Food Chem. 59 (2011)
[12] M. Ozyurek, N. Gungor, S. Baki, K. Guclu, R. Apak, Anal. Chem. 84 (2012) 12352–12360.
8052–8059. [59] B. Xu, S.K. Chang, J. Agric. Food Chem. 58 (2010) 1509–1517.
[13] M. Scampicchio, J. Wang, A.J. Blasco, A. Sanchez Arribas, S. Mannino, A. Escarpa, [60] L. Ziberna, M. Lunder, S. Moze, A. Vanzo, F. Tramer, S. Passamonti, G. Drevensek,
Anal. Chem. 78 (2006) 2060–2063. Cardiovasc. Toxicol. 10 (2010) 283–294.
[14] D. Vilela, M.C. Gonzalez, A. Escarpa, Anal. Bioanal. Chem. 404 (2012) 341–349. [61] J.K. Roy, L.R. Juneja, S. Isobe, T. Tsushida, Food Chem. 114 (2009) 263–269.
[15] D.P. Jones, Antioxid. Redox Signal. 8 (2006) 1865–1879. [62] C.B. Ahn, J.Y. Je, J. Food Biochem. 35 (2011) 1242–1256.
[16] E. Niki, Chem. Phys. Lipids 44 (1987) 227–253. [63] P.W. Buehler, A.I. Alayash, Antioxid. Redox Signal. 7 (2005) 1755–1760.
[17] G.R. Buettner, Arch. Biochem. Biophys. 300 (1993) 535–543. [64] N. Romero, A. Denicola, R. Radi, IUBMB Life 58 (2006) 572–580.
[18] V.E. Kagan, Y.Y. Tyurina, Ann. N.Y. Acad. Sci. 854 (1998) 425–434. [65] D. Honzel, S.G. Carter, K.A. Redman, A.G. Schauss, J.R. Endres, G.S. Jensen, J. Agric.
[19] E. Lonn, J. Bosch, S. Yusuf, P. Sheridan, J. Pogue, J.M. Arnold, C. Ross, A. Arnold, Food Chem. 56 (2008) 8319–8325.
P. Sleight, J. Probstfield, G.R. Dagenais, JAMA 293 (2005) 1338–1347. [66] M. Blasa, D. Angelino, L. Gennari, P. Ninfali, Food Chem. 125 (2011) 685–691.
[20] G.S. Omenn, G.E. Goodman, M.D. Thornquist, J. Balmes, M.R. Cullen, A. Glass, J.P. [67] I. Belinha, M.A. Amorim, P. Rodrigues, V. de Freitas, P. Moradas-Ferreira, N.
Keogh, F.L. Meyskens Jr., B. Valanis, J.H. Williams Jr., S. Barnhart, M.G. Cherniack, Mateus, V. Costa, J. Agric. Food Chem. 55 (2007) 2446–2451.
C.A. Brodkin, S. Hammar, J. Natl. Cancer Inst. 88 (1996) 1550–1559. [68] C. Dani, D. Bonatto, M. Salvador, M.D. Pereira, J.A. Henriques, E. Eleutherio, J.
[21] J.A. Scott, G.L. King, Ann. N.Y. Acad. Sci. 1031 (2004) 204–213. Agric. Food Chem. 56 (2008) 4268–4272.
[22] J.W. Finley, A.N. Kong, K.J. Hintze, E.H. Jeffery, L.L. Ji, X.G. Lei, J. Agric. Food Chem. [69] P.K. Wilmsen, D.S. Spada, M. Salvador, J. Agric. Food Chem. 53 (2005)
59 (2011) 6837–6846. 4757–4761.
[23] H.J. Forman, M. Maiorino, F. Ursini, Biochemistry 49 (2010) 835–842. [70] M.V. Baroni, R.D. Di Paola-Naranjo, C. García-Ferreyra, S. Otaiza, D.A. Wunderlin,
[24] A.L. Eggler, K.A. Gay, A.D. Mesecar, Mol. Nutr. Food Res. 52 (Suppl. 1) (2008) LWT – Food Sci. Technol. 47 (2012) 1–7.
S84–S94. [71] C.A. Stefenon, M. Colombo, C.M. Bonesi, V. Marzarotto, R. Vanderlinde, M. Sal-
[25] P. Renard, Y. Percherancier, M. Kroll, D. Thomas, J.L. Virelizier, F. Arenzana- vador, J.A.P. Henriques, Food Chem. 119 (2010) 12–18.
Seisdedos, F. Bachelerie, J. Biol. Chem. 275 (2000) 15193–15199. [72] J.D. Lambeth, Nat. Rev. Immunol. 4 (2004) 181–189.
[26] N.J. Miller, C. Rice-Evans, M.J. Davies, V. Gopinathan, A. Milner, Clin. Sci. (Lond.) [73] Y. Steffen, C. Gruber, T. Schewe, H. Sies, Arch. Biochem. Biophys. 469 (2008)
84 (1993) 407–412. 209–219.
[27] W. Brand-Williams, M.E. Cuvelier, C. Berset, LWT- Food Sci. Technol. 28 (1995) [74] Y. Steffen, T. Schewe, H. Sies, Biochem. Biophys. Res. Commun. 359 (2007)
25–30. 828–833.
[28] C. Henriquez, C. Aliaga, E. Lissi, Int. J. Chem. Kinet. 34 (2002) 659–665. [75] A. Agouni, A.H. Lagrue-Lak-Hal, H.A. Mostefai, A. Tesse, P. Mulder, P. Rouet, F.
[29] C. Sánchez-Moreno, J.A. Larrauri, F. Saura-Calixto, J. Sci. Food Agric. 76 (1998) Desmoulin, C. Heymes, M.C. Martinez, R. Andriantsitohaina, PLoS One 4 (2009)
270–276. e5557.
[30] V. Roginsky, E. Lissi, Food Chem. 92 (2005) 235–254. [76] R. Lopez-Sepulveda, M. Gomez-Guzman, M.J. Zarzuelo, M. Romero, M. Sanchez,
[31] I.F. Benzie, J.J. Strain, Anal. Biochem. 239 (1996) 70–76. A.M. Quintela, P. Galindo, F. O’Valle, J. Tamargo, F. Perez-Vizcaino, J. Duarte, R.
[32] R. Apak, K. Guclu, M. Ozyurek, S.E. Karademir, J. Agric. Food Chem. 52 (2004) Jimenez, Clin. Sci. (Lond.) 120 (2011) 321–333.
7970–7981. [77] T. Schewe, H. Kuhn, H. Sies, J. Nutr. 132 (2002) 1825–1829.
[33] N. Gungor, M. Ozyurek, K. Guclu, S.D. Cekic, R. Apak, Talanta 83 (2011) [78] V. Silva, G. Genta, M.N. Moller, M. Masner, L. Thomson, N. Romero, R. Radi, D.C.
1650–1658. Fernandes, F.R. Laurindo, H. Heinzen, W. Fierro, A. Denicola, J. Agric. Food Chem.
[34] L.M. Magalhaes, L. Barreiros, M.A. Maia, S. Reis, M.A. Segundo, Talanta 97 (2012) 59 (2011) 6430–6437.
473–483. [79] N. Wu, Y. Zu, Y. Fu, Y. Kong, J. Zhao, X. Li, J. Li, M. Wink, T. Efferth, J. Agric. Food
[35] E. Niki, Methods Enzymol. 186 (1990) 100–108. Chem. 58 (2010) 4737–4743.
[36] USDA, Database for the oxygen radical absorbance capacity (ORAC) [80] R.G. Knowles, S. Moncada, Biochem. J. 298 (1994) 249–258 (Pt 2).
of selected foods – Release 2, 2010 http://www.ars.usda.gov/Services/ [81] C. Auger, J.H. Kim, P. Chabert, M. Chaabi, E. Anselm, X. Lanciaux, A. Lobstein,
docs.htm?docid=1586 V.B. Schini-Kerth, Biochem. Biophys. Res. Commun. 393 (2010) 162–167.
[37] G. Cao, H.M. Alessio, R.G. Cutler, Free Radic. Biol. Med. 14 (1993) 303–311. [82] J.F. Leikert, T.R. Rathel, P. Wohlfart, V. Cheynier, A.M. Vollmar, V.M. Dirsch,
[38] B. Ou, M. Hampsch-Woodill, R.L. Prior, J. Agric. Food Chem. 49 (2001) Circulation 106 (2002) 1614–1617.
4619–4626. [83] S. Liu, J.E. Manson, I.M. Lee, S.R. Cole, C.H. Hennekens, W.C. Willett, J.E. Buring,
[39] A. Aspée, C. López-Alarcón, E. Lissi, Emerging Trends in Dietary Components Am. J. Clin. Nutr. 72 (2000) 922–928.
for Preventing and Combating Disease, in: B.S. Patil, G.K. Jayaprakasha, K.N.Ch. [84] Y. Takatsume, K. Maeta, S. Izawa, Y. Inoue, J. Agric. Food Chem. 53 (2005)
Murthy, N.P. Seeram (Eds.), ACS Symposium Series 1093 (2012) 417–429. 332–337.
[40] E. Alarcon, A.M. Campos, A.M. Edwards, E. Lissi, C. Lopez-Alarcon, Food Chem. [85] J. Fang, J. Lu, A. Holmgren, J. Biol. Chem. 280 (2005) 25284–25290.
107 (2008) 1114–1119. [86] J. Lu, L.V. Papp, J. Fang, S. Rodriguez-Nieto, B. Zhivotovsky, A. Holmgren, Cancer
[41] R.H. Bisby, R. Brooke, S. Navaratnam, Food Chem. 108 (2008) 1002–1007. Res. 66 (2006) 4410–4418.
[42] E. Nkhili, P. Brat, Anal. Bioanal. Chem. 400 (2011) 1451–1458. [87] Y. Wang, H. Zhang, A. Holmgren, W. Tian, L. Zhong, Oncol. Rep. 20 (2008)
[43] D.D. Wayner, G.W. Burton, K.U. Ingold, S. Locke, FEBS Lett. 187 (1985) 1479–1487.
33–37. [88] F. Arredondo, C. Echeverry, J.A. Abin-Carriquiry, F. Blasina, K. Antunez, D.P.
[44] E. Lissi, M. Salim-Hanna, C. Pascual, M.D. del Castillo, Free Radic. Biol. Med. 18 Jones, Y.M. Go, Y.L. Liang, F. Dajas, Free Radic. Biol. Med. 49 (2010) 738–747.
(1995) 153–158. [89] G. Shen, C. Xu, R. Hu, M.R. Jain, A. Gopalkrishnan, S. Nair, M.T. Huang, J.Y. Chan,
[45] A.M. Campos, C.P. Sotomayor, E. Pino, E. Lissi, Biol. Res. 37 (2004) 287–292. A.N. Kong, Mol. Cancer Ther. 5 (2006) 39–51.
[46] H. Puhl, G. Waeg, H. Esterbauer, Methods Enzymol. 233 (1994) 425–441. [90] S. Tanigawa, M. Fujii, D.X. Hou, Free Radic. Biol. Med. 42 (2007) 1690–1703.
[47] Q. Liu, H. Liu, Z. Yuan, D. Wei, Y. Ye, Colloids Surf. B: Biointerfaces 92 (2012) [91] E. Balogun, M. Hoque, P. Gong, E. Killeen, C.J. Green, R. Foresti, J. Alam, R. Mot-
348–352. terlini, Biochem. J. 371 (2003) 887–895.
[48] C.M. Lin, C.S. Chen, C.T. Chen, Y.C. Liang, J.K. Lin, Biochem. Biophys. Res. Com- [92] S.K. Biswas, D. McClure, L.A. Jimenez, I.L. Megson, I. Rahman, Antioxid. Redox
mun. 294 (2002) 167–172. Signal. 7 (2005) 32–41.
[49] M. Ozyurek, B. Bektasoglu, K. Guclu, R. Apak, Anal. Chim. Acta 636 (2009) 42–50. [93] C. Xie, J. Kang, M.E. Ferguson, S. Nagarajan, T.M. Badger, X. Wu, Mol. Nutr. Food
[50] C. Szabo, H. Ischiropoulos, R. Radi, Nat. Rev. Drug Discov. 6 (2007) 662–680. Res. 55 (2011) 1587–1591.
[51] P. Wardman, Radiat. Res. 170 (2008) 406–407. [94] K.L. Wolfe, R.H. Liu, J. Agric. Food Chem. 56 (2008) 8404–8411.
[52] A. Denicola, B. Alvarez, L. Thomson, in: P.E.S. Alvarez, A. Boveris (Eds.), Free
Radicals Pathophysiology, Transworld Research Network, Kerala, India, 2008,
pp. 39-55.

Potrebbero piacerti anche