Capacity Measurement. 1. Classification

Jan 4, 2016 - Antioxidant activity and inhibitory effect of melatonin and the relative indole compounds on perilla oil oxidation. Seok Joong Kim. Kore...
0 downloads 9 Views 1MB Size
Subscriber access provided by GAZI UNIV

Review

Antioxidant activity/capacity measurement: I. Classification, physicochemical principles, mechanisms and electron transfer (ET)-based assays Re#at Apak, Mustafa Özyürek, Kubilay Guclu, and Esra Capanoglu J. Agric. Food Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jafc.5b04739 • Publication Date (Web): 04 Jan 2016 Downloaded from http://pubs.acs.org on January 8, 2016

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Journal of Agricultural and Food Chemistry is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 105

Journal of Agricultural and Food Chemistry

Antioxidant activity/capacity measurement: I. Classification, physicochemical principles, mechanisms and electron transfer (ET)-based assays

Reşat Apak1, Mustafa Özyürek1, Kubilay Güçlü1, Esra Çapanoğlu2

1

Department of Chemistry, Faculty of Engineering, Istanbul University, Avcilar 34320,

Istanbul-Turkey

2

Department of Food Engineering, Faculty of Chemical and Metallurgical Engineering,

Istanbul Technical University, Maslak 34469, Istanbul-Turkey

* Corresponding Author. Tel.: +90 212 4737070 Fax: +90 212 473 7180 E-mail: [email protected]

1 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 2 of 105

ABSTRACT

1 2 3

Since there is no widely adopted “total antioxidant parameter” as a nutritional index for

4

labeling food and biological fluids, it is desirable to establish and standardize methods that

5

can measure the total antioxidant capacity (TAC) level directly from plant based food extracts

6

and biological fluids. In this review, we (i) present and classify the widely used analytical

7

approaches (e.g., in vitro and in vivo, enzymatic and non-enzymatic, electron transfer (ET)−

8

and hydrogen atom transfer (HAT)−based, direct and indirect assays) for evaluating

9

antioxidant capacity/activity; (ii) discuss total antioxidant capacity/activity assays in terms of

10

chemical kinetics and thermodynamics, reaction mechanisms, analytical performance

11

characteristics, together with advantages and drawbacks; (iii) critically evaluate electron

12

transfer (ET)-based methods for analytical, food chemical, biomedical/clinical and

13

environmental scientific communities so that they can effectively use these assays in their

14

correct places to meet their needs.

15 16

Keywords: Antioxidant activity; Total antioxidant capacity; Electron transfer-based

17

assays; Antioxidant mechanisms; Food analytical methods.

18 19

2 ACS Paragon Plus Environment

Page 3 of 105

20

Journal of Agricultural and Food Chemistry

1. INTRODUCTION

21 22

1.1. Scope

23 24

Oxidative stress is a pathological state in which reactive oxygen/nitrogen species (ROS/RNS)

25

overwhelm antioxidative defenses of the organism, leading to oxidative modification of

26

biological macromolecules (i.e., lipid, protein, DNA), tissue injury, and accelerated cellular

27

death1 as the foundation of many diseases. Measuring the antioxidant activity/capacity levels

28

of biological fluids and foods is carried out for the diagnosis and treatment of oxidative stress-

29

associated diseases in clinical biochemistry, for meaningful comparison of foods in regard to

30

their antioxidant content, and for controlling variations within or between products.

31

Antioxidant measurements have not yet been adapted to standard protocols in medical

32

diagnosis and treatment in spite of evidence-based changes of serum antioxidant capacity in

33

certain diseases (e.g., overall, total antioxidant capacity (TAC) of human serum was increased

34

in dialysis patients of chronic renal failure due to high urate values, but there was a marked

35

reduction after hemodialysis; antioxidant activity (AOA) of serum was markedly lower in

36

acute myocardial infarction and chronic lymphocytic leukaemia patients compared to those of

37

controls). So, ideally one should be able to detect/screen certain oxidative stress-originated

38

diseases and monitor the course of medical treatments by considering the changes in TAC

39

values of intracellular fluids and blood plasma/serum of a given individual measured by

40

standardized methods. The complexities of food and physiological applications of

41

antioxidants, separately and combined, require rigorous consideration and analysis of all

42

aspects of the chemistry, reaction mechanisms, and reaction/radical/target specificity in

43

various test systems, as well as careful and accurate quantitation of all reactants and products

44

involved. Current literature clearly state that there is no widely adopted “total antioxidant

3 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 4 of 105

45

parameter” as a nutritional index available for the labeling of food and biological fluids due to

46

the lack of standardized quantitation methods. Therefore it is desirable to establish and

47

standardize methods that can measure the TAC level directly from plant based food extracts

48

and biological fluids. Ideally, agreement on standardized methods for antioxidant testing (i)

49

requires general guidance for the application of assays, including exact description of test

50

conditions, experimental apparatus and stability of reagents; (ii) all new methods need to be

51

properly validated within their framework, associated with intra- and inter-laboratory

52

validation (including the production of repeatability, reproducibility and recovery data),

53

standardization, internal quality control, proficiency testing and analytical quality assurance;

54

(iii) the results should enable a reasonable comparison of the antioxidant content of foods,

55

pharmaceuticals and other commercial products; (iv) it may provide a measure for meeting

56

the need of quality standards for regulatory issues and health claims.2,3

57

To date, many in vitro tests are available from a chemical assay performed in a

58

homogenous solution to more complex methods using exogenic probes to detect oxidation.

59

Although many literature methods do not distinguish between ‘antioxidant capacity’ and

60

‘antioxidant activity’ of non-enzymatic antioxidants, end-point assays measuring the

61

efficiency of antioxidant action (i.e. reactive species inactivation) should be differentiated

62

from kinetic-based assays measuring reaction rate. TAC assays may be broadly classified as

63

electron transfer (ET)− and hydrogen atom transfer (HAT)−based assays, though in some

64

cases, these two mechanisms may not be differentiated with distinct boundaries (such as those

65

utilizing 2,2’-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid (ABTS) and 2,2-di(4-tert-

66

octylphenyl)-1-picrylhydrazyl (DPPH) radical reagents). In fact, most non-enzymatic

67

antioxidant activity (e.g., scavenging of free radicals, inhibition of lipid peroxidation, etc.) is

68

mediated by redox reactions. In addition to these two basic classes focusing on mechanism,

69

ROS/RNS scavenging assays will also be taken into account. Complementary to existing

4 ACS Paragon Plus Environment

Page 5 of 105

Journal of Agricultural and Food Chemistry

70

methods, novel approaches have recently been developed such as CUPric Reducing

71

Antioxidant Capacity (CUPRAC) TAC assay (introduced by our research group to world

72

literature in 2004), its modified ROS scavenging assays and other modifications (e.g.,

73

antioxidant sensor, postcolumn online HPLC technology). The current direction of CUPRAC

74

methodology can be best described as a self-sufficient and integrated train of measurements

75

providing a useful “antioxidant and antiradical assay package”.

76

The complexity and diversity of research topics investigated have led to the

77

development of a multitude of tests, but unfortunately none has gained universal acceptance.

78

Thus, one of the major challenges in antioxidant testing is to know which method is best

79

suited for a particular application. Since antioxidants may exert their effect through various

80

mechanisms such as scavenging radicals, sequestering transition metal ions, decomposing

81

hydrogen peroxide or hydroperoxides, quenching active prooxidants, and repairing biological

82

damage, it should be clarified which function of antioxidants is being measured, and

83

accordingly, the antioxidant assay method should be selected considering the function to be

84

evaluated.4 Because of the wide divergence of results for natural antioxidants in food systems,

85

more valid and rigorous guidelines and assay protocols are required to bring some order and

86

agreement to this important field. Naturally, it should be remembered that TAC and AOA are

87

not like elemental analysis parameters for which the analyst has to obtain more or less the

88

same result from different techniques (e.g., calcium in a milk sample measured by different

89

techniques should yield identical results within tolerable limits), because it is possible to get

90

quite different TAC or AOA results using the same probe under different experimental

91

conditions. Our understanding of the effects of antioxidant compounds can only be improved

92

with a deeper chemical insight if more specific methodology is used and is capable of

93

defining what products are formed and inhibited by antioxidants depending on conditions,

94

systems, and targets of protection.

5 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

95 96 97

Page 6 of 105

1.2. Technical Issues with TAC Assays

98 99 100

The technical issues associated with current ‘state-of-the-art’ TAC assays that require special consideration can be summarized as follows:

101

i. Almost total lack of standardization in experimental procedures and expressing results.

102

ii. Too many assays not having a demonstrated clear chemistry give rise to inconsistent

103

results, inappropriate application and interpretation of assays, and improper specification of

104

TAC. Naturally, statistical validation (e.g., using a certified reference material) of a given

105

assay is not possible in view of the different reaction kinetics and chemistry of the assays

106

producing different results.

107

iii. Most assays developed for TAC screening are devoid of detailed investigations related

108

to underlying initiators, targets, antioxidant interactions, kinetics, effects of solvent,

109

concentration and pH, etc. In earlier work regarding TAC assays, there seems to be some lack

110

of relevant classification according to chemical reaction mechanisms and kinetics. Molecular

111

accessibility problems of relevant reagents (such as steric hindrance, inner- and outer-sphere

112

electron transfer mechanisms of transition metal−based chromophores/fluorophores, etc.)

113

have not been discussed.

114

iv. Several studies on natural phytochemical compounds produced conflicting results

115

because of the non-specific “one-dimensional” character of methods used to evaluate

116

antioxidant activity. Since most natural antioxidants and phytochemicals are multifunctional

117

(i.e. due to variations in system composition, type of oxidizable substrate, media of initiation

118

and acceleration of oxidation, methods to assess oxidation and to quantify antioxidant

119

activity), a reliable antioxidant protocol requires the measurement of more than one property

120

relevant to either foods or biological systems.5

6 ACS Paragon Plus Environment

Page 7 of 105

Journal of Agricultural and Food Chemistry

121

v. A noteworthy question is that, if the protective action of an antioxidant is being

122

assayed in a selected test, what biomolecule (lipid, protein, DNA, etc.) is the antioxidant

123

supposed to protect, and how? Is the tested antioxidant at a significantly lower concentration

124

than that of the protected substrate? Are relevant ROS/RNS utilized in assaying the

125

antioxidant?6 In AOA and TAC assays covering a simulated oxidant-antioxidant reaction,

126

chromophore or luminescent probes capable of accepting H-atoms or electrons from

127

antioxidants do not necessarily protect relevant substrates (such as lipid, protein, DNA) from

128

oxidation, and therefore, these TAC assays results may not consequently reflect the capacity

129

to retard or suppress oxidation.7

130

vi. Antioxidant activity (i.e. related to the kinetics of antioxidant action for quenching

131

reactive species, usually expressed as reaction rates or scavenging percentages per unit time)

132

and antioxidant capacity (i.e. thermodynamic conversion efficiency of reactive species by

133

antioxidants, such as the number of moles of reactive species scavenged by one mole of

134

antioxidant during a fixed time period) are both important in antioxidant research, and care

135

must be exercised to distinguish between these two terms which are often used

136

interchangeably and therefore confused.8 Since the term ‘total antioxidant capacity’ (TAC) is

137

indicative of the collaborative (additive and possibly synergistic/antagonistic) action of all

138

antioxidants present in a complex sample, it is considered by most researchers as a more

139

useful parameter to assess the antioxidative defenses in food or plasma than separately

140

determining the concentrations of individual antioxidant constituents.

141

vii. Other than reactivity toward ROS/RNS, several factors such as concentration,

142

distribution, localization, fate of antioxidant−derived radical, interaction with other

143

antioxidants, and metabolism should be evaluated.4 The question of bioavailability and the

144

fate of metabolites of the antioxidant components (e.g., whether they undergo further redox

145

cycles) must be addressed in the case of in vivo assays.9 Also, in vitro antioxidant assays

7 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 8 of 105

146

carried out at unrealistic pH values (i.e. far from physiological pH, either in alkaline or acidic

147

range) cannot have much meaning for in vivo estimations of antioxidant action. In tests

148

carried out at weakly-acidic pH, most oxidation reactions for phenolic antioxidants are

149

incomplete within the protocol time of the assay.

150

viii. Due to the inadequacies in analytical methodology, the antioxidant activity of

151

proteins (i.e. first line defense elements against ROS attack in the human body) and

152

specifically of protein thiols are often ignored in most antioxidant assays, because proteins are

153

separated by precipitation from the main matrix and their contribution to serum TAC is left

154

unmeasured.10 Thus, the precise determination of serum TAC incorporating the “antioxidant

155

gap” originating from protein components is believed to be potentially useful to biochemists

156

investigating the diagnosis, treatment, prognosis, and follow-up of diseases utilizing TAC

157

measurement.11

158

ix.

Possible interactions of antioxidants among themselves (i.e. synergistic or

159

antagonistic effects) should be taken into account so as to better visualize the collaborative

160

action of antioxidants in the organism.

161

x.

Prooxidant effects of antioxidants, especially dependent on the composition of

162

medium in which antioxidant measurements are made, should also be considered, e.g.,

163

transition metal (especially iron) complex−based TAC assay reagents have the possibility of

164

redox cycling at their lower oxidation states that may falsify antioxidant test results.12

165

xi.

Antioxidant capacity assay results have not been effectively correlated to tests

166

measuring oxidative damage, such as thiobarbituric acid−reactive substances (TBARS) test in

167

lipids or carbonyl test in proteins. Normally, the difference in oxidative status of a medium

168

undergoing ROS/RNS attack, to which a food or biological extract was added, should

169

correlate to TAC of the extract under investigation.13 Additionally, antioxidant activity needs

170

to be correlated to electrochemical behaviour of antioxidants.

8 ACS Paragon Plus Environment

Page 9 of 105

Journal of Agricultural and Food Chemistry

171

As required parameters, a standardized TAC method; (i) utilizes a suitable oxidant

172

which actually is or a simulator of a biologically relevant reactive species; (ii) is simple,

173

practical and versatile; (iii) uses a method with a defined end-point and a clear chemical

174

mechanism; (iv) has readily available and preferably low-cost instrumentation; (v) is

175

reproducible with good within-run and between-run precision; (vi) can assay both hydrophilic

176

and lipophilic antioxidants; (vii) does not generate new reactive species that may cause under-

177

or over-estimated TAC readings; (viii) is suitable for “high-throughput” analysis for routine

178

quality control of food and biological extracts.2,14

179 180

1.3. Purpose

181 182

The aims of these comprehensive review series are; (i) to present a brief panorama of the most

183

widely used methods and of new analytical approaches for evaluating antioxidant

184

capacity/activity; (ii) to discuss TAC/AOA assays in terms of chemical kinetics and

185

thermodynamics, reaction mechanisms, analytical performance characteristics (linear

186

concentration range, recovery, repeatability, reproducibility, and recognition of interfering

187

substances, etc.), and advantages and drawbacks; (iii) to bring in terms of definitions or

188

definition-like characterization and classification of the chemical and biochemical methods of

189

antioxidant assays as well as related antioxidant chemistry; and finally (iv) to provide a

190

critical evaluation of this topic to analytical, food chemical, biomedical/clinical and

191

environmental scientific communities so that they can effectively use these assays in their

192

correct places to meet their needs. The literature gap summarized in the above ‘Technical

193

issues with TAC assays’ is endeavoured to be filled. The basic criteria of classification of

194

antioxidant assays, such as in vitro and in vivo, enzymatic and non-enzymatic, electron

195

transfer (ET)− and hydrogen atom transfer (HAT)−based, direct and indirect assays have been

9 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 10 of 105

196

addressed. The essential ET−based assays with possible advantages/drawbacks have been

197

critically evaluated, and at the same time, various methodologies of the main and modified

198

CUPRAC procedures regarding TAC and ROS/RNS-scavenging assays have been unified and

199

summarized. However in the first review of this series, the stress will be upon ET assays, and

200

HAT and mixed-mode (ET/HAT) assays, together with lipid peroxidation assays, ROS/RNS

201

scavenging assays and oxidative stress biomarkers are the subjects of the following reviews.

202 203 204

2. CLASSIFICATION OF AOA AND TAC ASSAYS

205 206

‘Antioxidants’ are natural or synthetic substances that may prevent or delay oxidative cell

207

damage caused by physiological ‘oxidants’ having distinctly positive reduction potentials,

208

covering ROS/RNS and free radicals (i.e. unstable molecules or ions having unpaired

209

electrons). The terms ‘oxidant’ and ‘antioxidant’ have complementary meanings in the sense

210

that these compounds neutralize the effects of each other. On the other hand, a ‘prooxidant’

211

may not have a large reduction potential by itself, but may induce oxidative damage to

212

various biological targets like DNA (e.g., nucleic base modification and single/double strand

213

breaks), lipids (e.g., structural changes in fatty acid composition and lipid peroxidation) and

214

proteins (e.g., protein carbonylation and oxidation of certain amino acid moieties).15 For

215

example, transition metal ions at their lower oxidation states are not oxidant species by

216

themselves, but may induce the generation of ROS/RNS with hydrogen peroxide or molecular

217

oxygen, thereby acting as prooxidants. Antioxidants may be broadly defined as ‘substances

218

that, when present at relatively low concentrations compared with those of the oxidizable

219

substrates, significantly delay or inhibit oxidation of those substrates’.16,17 Although the term

220

‘oxidizable substrate’ includes every type of molecule found in vivo, it is generally

221

understood as biomacromolecules like lipid, protein and DNA. This definition emphasizes the

10 ACS Paragon Plus Environment

Page 11 of 105

Journal of Agricultural and Food Chemistry

222

importance of the selected damage target and the source of ROS/RNS used when antioxidants

223

are tested and their actions are examined.18 However, Finley et al.19 points out to the

224

ambiguity of this definition in that the term “antioxidant” may have different connotations to

225

different audiences, such as the capability of quenching metabolically generated ROS (to

226

biochemists and nutritionists), the functionality of retarding food oxidation (to food

227

scientists), or the property of yielding high TAC values in ET− and HAT−based in vitro

228

assays (presumably to a wider community of food science, commerce and industry). For

229

convenience, antioxidants have been traditionally divided into two classes; primary or chain-

230

breaking antioxidants (mainly acting by ROS/RNS scavenging), and secondary or

231

preventative antioxidants (usually acting by transition metal ion chelation).20 Thus, an

232

antioxidant may act directly by scavenging reactive species, or inhibiting their generation. It

233

may also act indirectly, e.g., by up-regulating endogenous antioxidant defenses.6,21 This work

234

is concerned with the corresponding methods of antioxidant capacity/activity assay capable of

235

measuring chain-breaking or preventive antioxidant ability. In lipid peroxidation experiments,

236

antioxidants acting only by metal chelation are essentially maintained, whereas chain-

237

breaking antioxidants are consumed. Most of the time, this difference is reflected in a lag

238

(retardation) phase of the peroxidation process by chain-breaking antioxidants, compared to a

239

constant inhibition by metal-binding antioxidants throughout the reaction.6,21 However, the

240

scope of this review is limited to the integrated activity of non-enzymatic antioxidants,

241

meaning that endogenous antioxidative enzymes such as superoxide dismutase (scavenging

242

superoxide anion radicals), catalase and glutathione peroxidase (able to remove hydrogen

243

peroxide) are not considered.

244

Chain-breaking mechanisms regarding the breaking of the oxidation chain of lipid

245

radicals (L•, LOO•, or LO•) involve the sacrificial consumption of antioxidants (AH) to

246

produce antioxidant radicals (A•) protecting lipid molecules (L):

11 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

247

L• + AH → LH + A• … (Eq. 1)

248

LOO• + AH → LOOH + A•. … (Eq. 2)

249

LO• + AH → LOH + A• … (Eq. 3)

Page 12 of 105

250

Thus, radical initiation (by reacting with a lipid radical) or propagation (by reacting

251

with lipid peroxyl or alkoxyl radicals) steps are inhibited. Since chain-breaking antioxidants

252

exert their action through either hydrogen atom (H•) and electron (e-) donation or both (i.e.

253

proton-coupled electron transfer), such AOA measurement methods are commonly classified

254

as hydrogen atom transfer (HAT)− and electron transfer (ET)−based assays according to

255

mechanism.

256

On the other hand, secondary (or preventive) antioxidants retard or prevent lipid

257

oxidation. For example, transition metal ion (e.g., Fe(II) or Cu(I)) chelator antioxidants may

258

inhibit Fenton-type reactions that produce hydroxyl radicals which may cause oxidative

259

degradation of biological macromolecules (lipids, proteins, DNA, etc.):

260

Fe(II) + H2O2 → Fe(III) + •OH + OH- … (Eq. 4)

261

Cu(I) + H2O2 → Cu(II) + •OH + OH- … (Eq. 5)

262 263

Therefore, preventive antioxidant assay methods should measure transition metal ion chelating ability.

264

The transition metal chelation functionality of antioxidants covers a neutralization

265

reaction between a Lewis base (antioxidant) and a Lewis acid (metal ion), without involving

266

the donation of H-atoms or electrons by the antioxidant. Therefore, the measurement of such

267

preventive AOA should be covered under a different category. On the other hand, hindering

268

the formation of ROS/RNS should be considered as a preventive action,7 while scavenging of

269

ROS/RNS is closely related to HAT and ET mechanisms for the measurement of chain-

270

breaking AOA. Nevertheless, techniques for the measurement of scavenging activity of

271

reactive species (e.g., ROS essentially cover hydroxyl radical: •OH, superoxide anion radical:

12 ACS Paragon Plus Environment

Page 13 of 105

Journal of Agricultural and Food Chemistry

272

O2•-, singlet oxygen: 1O2, hydrogen peroxide: H2O2, and hypochlorous acid (HOCl); RNS

273

mainly cover peroxynitrite: ONOO-, and nitric oxide: •NO) are investigated under a separate

274

subtitle.

275

Enzymatic antioxidants are either reductase enzymes (e.g., superoxide dismutase

276

(SOD), glutathione peroxidase (GSH-Px), glutathione reductase (GSH-Rx), and catalase) and

277

their cofactors, which limit the cellular concentration of free radicals and prevent excessive

278

oxidative damage, or oxidase enzyme inhibitors. Therefore, the corresponding enzymatic

279

AOA assays should either measure the enzymatic reduction ability of ROS or the inhibition of

280

oxidases (e.g., xanthine oxidase, NADPH oxidase) capable of producing reactive species. On

281

the other hand, non-enzymatic AOA/TAC assays utilize a relevant probe for simulating the

282

antioxidative action toward oxidant species. Since antioxidant and oxidant-regenerating

283

enzymes in blood cells and the blood vessel wall have a profound impact on the antioxidant

284

properties of blood plasma (which is not reflected in the in vitro assays of isolated plasma),

285

the term ‘TAC’ has been claimed to measure only a part of antioxidant capacity, usually

286

excluding enzymatic activities, and therefore 'non-enzymatic antioxidant capacity' (NEAC)

287

has been suggested as a more relevant term than TAC.22 For example, non-enzymatic plasma

288

antioxidants usually cover albumin and other related proteins containing thiols and other

289

antioxidative amino acid residues, as well as small molecules like α-tocopherol, bilirubin,

290

ascorbic acid, uric acid, and reduced glutathione (GSH). In the literature, much more effort

291

has been spent on developing non-enzymatic antioxidant assays covering a wide range of

292

HAT− and ET−based assays, and methods for measuring ROS/RNS scavenging activity.

293

Recently, synthetic and natural phenolic antioxidants have been summarized, together with

294

their mode of action, health effects, degradation products and toxicology.23 A general and

295

update overview of methods available for measuring antioxidant activity and the chemistry

13 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 14 of 105

296

behind them has been provided.24 Advantages and limitations of common testing methods

297

have been listed, with a preference for method selection serving different needs.25

298 299

2.1. Measurement of non-Enzymatic Chain-Breaking Antioxidant Activity/Capacity

300 301

Non-enzymatic chain-breaking antioxidant ability can be measured by finding the rate (kinetic

302

AOA methods) or thermodynamic conversion efficiency (TAC methods) for the reaction of a

303

suitable oxidant probe with the antioxidant. AOA assays like TRAP (total peroxyl radical

304

trapping antioxidant parameter),10,26 crocin bleaching,27,28 ORAC (oxygen radical absorbance

305

capacity),29,30 TOSC (total oxyradical scavenging capacity),31,32 etc. are usually competitive

306

(Figure 1) and work on a HAT mechanism, whereas TAC measurement methods are usually

307

non-competitive (Figure 2) ET and mixed-mode (ET/HAT) assays. In a competitive

308

inhibition (or scavenging) assay, the oxidant reacts with a probe leading to changes in its

309

absorbance, fluorescence, luminescence, or any other measurable property, where

310

antioxidants compete with the probe for the oxidant and repair the oxidized probe.33 Due to

311

the competition between the probe and antioxidants for reactive species, the probe undergoes

312

less oxidative conversion by ROS/RNS in the presence of antioxidants. A major criticism

313

directed at HAT-based competitive assays using fluorescent probes is that the concentration

314

of the target species (i.e. assay probe simulating a biological substrate) is usually smaller than

315

that of tested antioxidants,14 which contradicts with the basic ‘definition of antioxidant’,

316

because in real life, antioxidants exert their protective effects even when they are at much

317

lower concentration than that of the biological (oxidizable) substrate.15 On the other hand, in

318

ET−based assays, the probe undergoing reduction with the antioxidant is either converted to a

319

colored, fluorescent, chemiluminescent etc. species, or the initial absorbance/fluorescence is

320

attenuated as a result of the antioxidation reaction. ET−based assays have been criticised

14 ACS Paragon Plus Environment

Page 15 of 105

Journal of Agricultural and Food Chemistry

321

mainly because the utilized probe acting as the oxidizing agent does not involve

322

physiologically important oxidants like ROS/RNS, causing a discrepancy between the

323

simulated assay and the real-life antioxidant action.

324

Figure 1

325

Figure 2

326 327

The most widely used chromophores in ET−based spectrophotometric TAC assays of (2,2’-azino-bis(3-ethylbenzothiazoline-6-sulfonic

328

Folin-Ciocalteu,34,35

329

acid/Trolox®-equivalent antioxidant capacity),36,37 DPPH (2,2-di(4-tert-octylphenyl)-1-

330

picrylhydrazyl),38,39 CUPRAC (cupric reducing antioxidant capacity),40,41 FRAP (ferric

331

reducing antioxidant power),42-45 ferricyanide,46,47 ferric-phenanthroline,48 and ferric-

332

ferrozine49 assays are phospho-tungsto-molybdate(V), ABTS•+ radical cation, DPPH• radical,

333

cuprous neocuproine: [Cu(Nc)2]+ chelate, ferrous tripyridyltriazine: [Fe(TPTZ)3]2+ chelate,

334

Prussian blue: K[Fe(Fe(CN)6] heteropoly acid salt, ferrous phenanthroline: [Fe(phen)3]2+

335

chelate, and [Fe(FZ)3]2+ chelate, respectively. When transition metal ion−based probes such

336

as ferric and cupric chelates oxidize the to-be-assayed antioxidant compounds, the metal ion

337

placed at the coordination center of the complex is reduced to the lower oxidation number and

338

its chelate has strong intermolecular charge-transfer interactions, i.e. visible light absorbed by

339

the chelate causes a partial transfer of electronic charge from the metal ion to the ligand

340

(giving rise to very high molar extinction coefficients for these chromophores, thereby raising

341

the sensitivity for antioxidant determinations). Some authors classify DPPH and ABTS tests

342

as mixed-mode (i.e. having both ET- and HAT mechanisms) assays.2 TAC assays using

343

Cr(VI) as chromate50 and Ce(IV)51-53 as oxidant are also ET-based methods where

344

antioxidants reduce these species to Cr(III) and Ce(III), respectively; naturally, certain

345

measures have to be taken to decrease the oxidizing ability of these reagents so that

ABTS/TEAC

15 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 16 of 105

346

antioxidants but not other organic compounds (e.g., citric acid and common sugars in food

347

and beverages) are oxidized in the assays. When gold nanoparticles (Au-NPs) generated from

348

HAuCl4 upon reduction with phenolic acid antioxidants are used as colored probes exhibiting

349

localized surface plasmon resonance (LSPR) absorption, the highest capacity of reducing

350

gold(III) to elemental gold nanoparticles corresponds to the highest antioxidant activity,

351

consistent with the tendency of phenolic antioxidants to donate electrons.54 The same

352

reasoning applies for the formation of silver nanoparticles (Ag-NPs) coatings generated from

353

AgNO3 with phenolic antioxidants onto citrate-stabilized silver seeds, where the enlargement

354

of preformed Ag-NPs gave rise to enhanced LSPR absorption linearly dependent on

355

polyphenol concentration.55

356 357

2.1.1. HAT−Based Methods

358 359

HAT−based assays measure the capability of an antioxidant to quench free radicals (generally

360

peroxyl radicals) by H-atom donation. Peroxyl radicals are generally chosen as the reactive

361

species in these assays because of their higher biological relevance and longer half-life

362

(compared to hydroxyl and superoxide radicals). The HAT mechanism of antioxidant action,

363

in which the hydrogen atom (H•) of a phenol (Ar-OH) is transfered to an ROO• radical, can be

364

summarized by the reaction:

365

ROO• + AH/ArOH → ROOH + A• / ArO• … (Eq. 6)

366

where the aryloxy radical (ArO•) formed from the reaction of antioxidant phenol with peroxyl

367

radical is usually stabilized by resonance. The AH and ArOH species denote the protected

368

biomolecules and antioxidants, respectively. Effective phenolic antioxidants need to react

369

faster than biomolecules with free radicals to protect the latter from oxidation.56 As major

370

criticisms directed at HAT–based assays having a ‘lag-phase’ approach (i.e. measuring the

371

retardation time for the initiation of oxidative probe conversion) to quantification of 16 ACS Paragon Plus Environment

Page 17 of 105

Journal of Agricultural and Food Chemistry

372

antioxidant capacity; (i) not every antioxidant possesses an apparent lag-phase (i.e. in order to

373

have a distinct lag-time, an antioxidant should have a rate constant for the tested radical much

374

higher than that of the probe such that the antioxidant should be consumed up by the time

375

when the probe oxidation is observable),22 (ii) ambiguity in end-point observation makes

376

inter-laboratory comparison of generated data difficult, and (iii) the antioxidant capacity

377

profile of samples following the lag-phase is disregarded.15 Since in HAT–based antioxidant

378

assays using a fluorescent probe, both the probe and antioxidants react with ROO•, the

379

antioxidant activity can be determined from competition kinetics (Figure 1) by measuring the

380

fluorescence decay curve of the probe in the absence and presence of antioxidants, and

381

integrating the area under these curves (AUC approach). The AUC difference between a

382

sample and a reagent blank is then related to antioxidant concentration in the sample.2,14

383

2.1.2. ET−Based Methods

384 385

The ET mechanism of antioxidant action with a biologically relevant radical is based on the

386

reactions:

387 388 389 390 391 392 393

ROO• + AH/ArOH → ROO- + AH•+/ArOH•+ … (Eq. 13)

394

assays (though for metal-complex probes capable of outer-sphere electron transfer, this

395

assumption was shown to be invalid as a function of solvent type), and are both solvent– and

396

pH–dependent. The pH-dependency is apparent from the above reaction sequence of ET

397

mechanism. For example, phenolic compounds (Ar-OH) having weakly acidic -OH groups

398

dissociate to a greater extent at higher pH, and become more susceptible to oxidation. Thus,

399

most ET reactions occur at a higher rate at higher pH. The reactivity of the ABTS•+ cation

AH•+/ArOH•+ + H2O ↔ A• / ArO• + H3O+

… (Eq. 14)

ROO- + H3O+ ↔ ROOH + H2O … (Eq. 15) where the reactions are generally assumed to be relatively slower than those of HAT– based

17 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 18 of 105

400

radical toward ascorbic acid at neutral pH is expressed with a second-order rate constant of

401

8x106 M-1s-1, whereas in acidic pH, this rate constant decreases by almost two orders-of-

402

magnitude.57 Generally, ionization potentials of phenolic antioxidants decrease with

403

increasing pH, which causes an increase in electron-donating capacity concomitant with

404

deprotonation.2 Iron(III)-based antioxidant assays conforming to ET mechanism (excluding

405

the sole hexacyanoferrate(III) complex not in combination with ferric ion) have to be carried

406

out at an acidic pH in order to prevent the hydrolysis of trivalent iron (e.g., giving rise to

407

FeOH2+ and further hydrolysis complexes).48 The aryloxy radical (ArO•) is subsequently

408

oxidized to the corresponding quinone (Ar=O). The more stabilized the aryloxy radical is, the

409

easier will be the oxidation from ArOH to Ar=O due to reduced redox potential,56 and the

410

stronger is the antioxidant.

411

Electron transfer (ET)‒based (or reduction‒based) assays can be better understood

412

considering the fact that antioxidants are also good reducing agents capable of reductive

413

quenching of ROS/RNS. However, these assays do not necessarily use biologically relevant

414

reactive species (such as peroxyl radicals); instead they use artificial probes that change color

415

or fluorescence when reduced by antioxidants (Figure 2).14,56 For example, the cupric- or

416

ferric-reducing ability measured for a biological sample may indirectly but efficiently reflect

417

the total antioxidant power of the sample even though no radical species are involved in the

418

assay. The change in absorbance or fluorescence of the probe (at a pre-specified wavelength)

419

upon reduction with antioxidants is a measure of the total concentration of antioxidants in a

420

sample, or TAC. This TAC is usually expressed in terms of a reference compound, such as

421

Trolox for hydrophilic antioxidants, α-tocopherol (vitamin E) for lipophilic antioxidants, and

422

gallic acid for aqueous solutions of polyphenols, where trolox-equivalent and gallic acid-

423

equivalent TAC values are denoted as TE and GAE, respectively. The TEAC coefficient is a

424

unitless value, defined as the reducing potency −in Trolox® mM equivalents− of 1 mM

18 ACS Paragon Plus Environment

Page 19 of 105

Journal of Agricultural and Food Chemistry

425

antioxidant solution under investigation. Usually in spectrophotometric TAC assays, this

426

TEAC coefficient is found from the ratio of the slope of the calibration curve (drawn as

427

absorbance versus concentration) of the tested compound to that of Trolox obtained under

428

identical conditions. The TAC value, in mM-TE units, of a complex antioxidant mixture

429

(comprised of antioxidants: 1,2,3,…, i, …n) is the sum of the products obtained by

430

multiplying the mM concentration (Ci) of each antioxidant with its TEAC coefficient

431

(TEACi): TAC(mixture) = ΣCi(TEAC)i

432

Naturally this equation is valid as long as the principle of additivity of absorbances is

433

retained for a complex mixture conforming to Beer’s law of spectrophotometry, i.e. the TAC

434

of a complex antioxidant mixture is the sum of the TAC values of individual antioxidants

435

comprising the mixture.

436

In general, ET–based TAC assays have good precision, because the difference in

437

absorbance or fluorescence intensities of the reduced and original probes can be directly

438

measured. Since most of the time a single reduced species is produced from the probe upon

439

chemical reduction with antioxidants, the absorbance changes of a single chromogenic

440

product at a fixed pH and wavelength usually vary almost perfectly linearly with total

441

antioxidant concentration (in TE units). Thus, within the linear concentration range obeying

442

Beer’s law, additivity of absorbances (and therefore additivity of TAC values of individual

443

constituents forming an antioxidant mixture) is usually attained. Reduction–based assays have

444

generally been criticised for not involving biologically relevant radicals. However, their

445

simulated conditions can well imitate the media in food and biological fluids such as redox

446

potential, pH, and lipophilic/hydrophilic balance of solvent mixtures and microemulsions.

447

With the exception of the Folin-Ciocalteu reagent having an indefinite redox potential, most

448

ET reagents have standard potentials in the useful range of 0.6-0.7 V relevant for food and

449

biological fluids, i.e. they can oxidize important antioxidants to measure their TAC values.

19 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 20 of 105

450

The reaction conditions of reduction-based TAC assays should be well defined so as to

451

maintain reproducibility, since inter-assay results without detailed description of conditions

452

cannot be compared. The time and temperature of measurements should always be indicated

453

in regard to repeatability of reaction kinetics, because some oxidation reactions of ET probes

454

with antioxidants may not reach saturation within the pre-specified protocol time of the

455

assays. For example, high-spin Fe(III) having half-filled d-orbitals may show a kinetic

456

inertness to thiols, some phenolic acids and flavonoids such that the envisaged oxidation

457

cannot be completed within the protocol time period of the FRAP assay and similar Fe(III)-

458

based assays.48

459

When ET-based reagents, especially metal complexes utilizing Fe(III) or Cu(II), are to

460

be used in the TAC determination of plasma or serum, it should be remembered that these

461

samples should be preserved in the cold with either heparin or citrate and not EDTA, because

462

EDTA usually stabilizes the higher oxidation state (such as Fe(III) of Cu(II)) in preference to

463

the lower one (e.g., Fe(II) or Cu(I)) by forming a more stable complex, and this decreases the

464

Nernst potential of the concerned redox couple in the presence of a chromogenic ligand (e.g.,

465

tripyridyltriazine or neocuproine), thereby weakening the oxidizing power of the reagent

466

against certain plasma antioxidants and causing negative errors in TAC measurement. Bartosz

467

recommended the use of human plasma in TAC studies rather than serum, which should be

468

analyzed immediately after blood collection; he also recommended the use of citrate or

469

heparin rather than EDTA as anticoagulant for serum samples (EDTA may only be

470

permissible at low concentrations), and pointed out to cases where serum yielded higher TAC

471

values than plasma due to the possible release of antioxidants from blood platelets during the

472

clotting process.33

473

There is lack of correlation between activities determined by the same antioxidant by

474

different assays and between activities determined by the same assay in different

20 ACS Paragon Plus Environment

Page 21 of 105

Journal of Agricultural and Food Chemistry

475

laboratories.58 Especially assays based on the inhibition of lipid peroxidation are known to

476

correlate poorly with either HAT- or ET-based assays, meaning that an antioxidant with a

477

high TEAC value in routine TAC assays may not perform well in preventing/retarding lipid

478

peroxidation. When the TAC results found by different methods for complex samples are

479

compared, there may only be a good linear correlation (i.e. r2 ≈ 1) but not a one-to-one

480

identity between them. The reason is that every assay has its own unique thermodynamic and

481

kinetic characteristics, and the oxidizing power of each TAC reagent against a given

482

antioxidant within a fixed time is naturally different from one another. Even the same assay

483

with slight differences in reagent preparation may give rise to serious differences in results:

484

for example, ORAC assay using two different probes, β-phycoerythrin and fluorescein, may

485

give entirely different TEAC values for quercetin as 2.07±0.05 and 7.28±0.22, respectively;

486

ABTS radical yields 0.55 and 0.94 TEAC values for glutathione using the ABTS/H2O2/HRP

487

and ABTS•+ decolorization methods, respectively; the TEAC value for ascorbic acid may

488

change three-fold depending on the type of oxidant used for generating ABTS•+ radical, i.e.

489

MnO2 versus persulfate.33 Generally ET-based assays correlate well among themselves due to

490

the similarity in mechanism of action, and therefore some antioxidant researchers consider the

491

application of a series of ET-based assays redundant. On the other hand, it is well known that

492

HAT- and ET-based assays like ORAC, ABTS/TEAC and FRAP give none or weak

493

correlations for plasma samples.33 In this regard, the antioxidant activities of common

494

vegetables (total sample size: 927) collected from the U.S. market, analyzed using the ORAC

495

and FRAP procedures, did not correlate well.59 Cao and Prior observed a weak linear

496

correlation between serum ORAC and serum FRAP, but no correlation either between serum

497

ORAC and serum ABTS/TEAC, or between serum FRAP and serum ABTS/TEAC.60 Prior et

498

al. recommended the evaluation of overall antioxidant capacity by using multiple assays to

499

generate an “antioxidant profile” encompassing reactivity toward both aqueous and

21 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 22 of 105

500

lipid/organic radicals directly via radical quenching and radical reducing mechanisms and

501

indirectly via metal complexing.2 It may generally be recommended to use a variety of assays

502

with different mechanisms (such as ET-, HAT- and lipid peroxidation-based assays) for

503

complex samples in order to see the whole picture for antioxidant action.

504 505

2.1.2.1. Spectroscopic Methods

506 507

2.1.2.1.1. Folin-Ciocalteu (FC) Assay

508 509

The Folin−Ciocalteu (FC) method was initially intended for the analysis of proteins, taking

510

advantage of the reagent’s activity toward protein tyrosine (containing a phenol group)

511

residue, where tyrosine reacted with the heteropoly reagent to give a blue color proportionate

512

to the protein content.34 Much later, Singleton et al.35 extended this assay to the analysis of

513

total phenols in wine. Fundamentally, the FC assay is based on the oxidation of phenol

514

compounds in alkaline (carbonate) solution with a molybdotungstophosphate heteropolyanion

515

reagent (3H2O-P2O5-13WO3-5MoO3-10H2O), yielding a colored product with a broad band

516

having an absorbance maximum (λmax) laying between 750 and 765 nm. Although a similar

517

phosphomolybdenum blue formation method, i.e. without tungstate in the reagent, was also

518

reported for antioxidant capacity determination in acidic medium at elevated temperature,61 it

519

was not tested for a wide variety of antioxidants and its molar absorptivity for vitamin E was

520

rather low, which possibly restricted further practice of the method.

521

The possible simultaneous occurrence of several reduced species in the FC heteropoly-

522

chromophore can account for the broad peaks. Although the color may be developed more

523

quickly at warmer temperature, the loss of color with time is greater at higher temperatures.

524

As tannic acid from different preparations of wines and spirits could vary, and other tannins

22 ACS Paragon Plus Environment

Page 23 of 105

Journal of Agricultural and Food Chemistry

525

covered a wide range of color yield per unit weight, Singleton et al.35 replaced tannin (as a

526

reference compound) with GAE in reporting FC results; the minimum detectable amount of

527

phenols was at the order of 3 mg GAE/liter, depending on optical cuvette thickness. The

528

gallic acid added to wine was recovered quantitatively, and the absorbance produced from a

529

mixture of natural phenols of different classes was equivalent to the sum of their individual

530

contributions, meaning that chemical deviations from Beer’s law was essentially absent in the

531

FC system.

532

Neither the exact chemical nature nor the redox potential of the FC phenol reagent is

533

definitely known. It is a strong oxidizing agent, and can non-specifically oxidize many non-

534

phenolic reducing compounds including weak reductants (e.g., aromatic amines, sulphite,

535

ascorbic acid, Cu(I), Fe(II), etc.) along with phenolics.15 Due to this oxidative ability, FC was

536

proposed to be used as a TAC reagent in the reducing capacity assay for antioxidants, where

537

the molybdenum center in the complex reagent is reduced from Mo(VI) to Mo(V) with an

538

electron donated by an antioxidant to produce a blue color.14 Unfortunately, the detailed

539

molecular and electronic structures of the blue reduction products are unclear, but it is known

540

that molybdates are more easily reduced than tungstates in heteropoly salts. FC assay has

541

certain advantages over some other TAC assays in that it is simple, fast, robust, does not

542

require specialized equipment, and the long-wavelength absorption of the chromophere

543

minimizes interference from the sample matrix. However, a drawback of the FC assay is that

544

reducing agents such as ascorbic acid, citric acid, simple sugars or certain amino-acids can

545

interfere with the analysis and thus overestimate the content of phenolic compounds.

546

Tryptophan, indoles, purines, guanine, xanthine, and uric acid were also reported to react with

547

the FC reagent to yield molybdenum blue.35 Another disadvantage is that its commercially

548

available reagent is to be used for reproducible TAC assays, since its preparation by separate

549

laboratories is rather cumbersome. The conventional FC reagent is only applicable to water-

23 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 24 of 105

550

soluble antioxidants, and operates at an unrealistically high pH. To avoid the possible air

551

oxidation of the tested phenols before the color reaction, the FC reagent should be added

552

before alkali. Nevertheless, FC is routinely practiced in antioxidant research laboratories for

553

testing food and plant extracts. Its fully automated-continuous flow 40-sample/hour procedure

554

was also adapted.62

555

Since most phenolic compounds are in dissociated form (as conjugate bases, mainly

556

phenolate anions) at the working pH of the assay (pH ≈10), they can be more easily oxidized

557

with the FC reagent, possibly giving rise to an overestimated TAC value.14,56 The FC

558

chromophore, the molybdotungstophosphate heteropolyanion (PMoW11O404-), does not have

559

an affinity toward organic solvents owing to its quadruple negative charge2 giving rise to

560

strong ion−dipole interactions with solvent water molecules. Thus, the FC method was

561

modified and standardized by Berker et al.63 so as to enable simultaneous measurement of

562

lipophilic and hydrophilic antioxidants in NaOH-added isobutanol−water medium. The

563

modified procedure was successfully applied to the total antioxidant capacity assay of Trolox,

564

quercetin, ascorbic acid, gallic acid, catechin, caffeic acid, ferulic acid, rosmarinic acid,

565

glutathione, and cysteine, as well as of lipophilic antioxidants such as α-tocopherol (vitamin

566

E), butylated hydroxyanisole, butylated hydroxytoluene, tert-butylhydroquinone, lauryl

567

gallate, and β-carotene.63

568 569

2.1.2.1.2. FRAP Assay

570 571

The FRAP assay, first introduced by Benzie and Strain to determine the antioxidant capacity

572

of plasma and later modified for application to other matrices such as tea and wine,42-45 is

573

based on the reduction of Fe(III) to Fe(II) by antioxidants in the presence of tripyridyltriazine

574

tridentate ligand forming a colored complex with Fe(II):

24 ACS Paragon Plus Environment

Page 25 of 105

575 576 577 578

Journal of Agricultural and Food Chemistry

Fe(TPTZ)23+ + ArOH → Fe(TPTZ)22+ + ArO• + H+ … (Eq. 16) where TPTZ denotes 2,4,6-tripyridyl-s-triazine ligand, and the absorption maximum lies at a

579

wavelength of λmax = 593 nm. Although Fe(III)-Fe(II) reduction may equally well produce

580

colored chelates in the presence of ortho- and batho-phenanthrolines yielding reduction

581

potentials exceeding 1.0 V, TPTZ was carefully selected, because EoFe(III)-Fe(II) in the presence

582

of TPTZ is only 0.70 V which may selectively oxidize most antioxidants but not citric acid

583

and simple sugars. The FRAP assay is simple, practical, inexpensive, and may offer a putative

584

index of antioxidant capacity.15 Since the FRAP reaction with antioxidants produces a single

585

colored product, i.e. Fe(II)-TPTZ, the FRAP absorbance versus concentration curves are well

586

linear over a wide range, and TAC additivity is usually observed in mixtures ‒ except for

587

those containing small-molecular and protein thiols.11,64

588

Pulido et al.45 compared the antioxidant efficiency of a number of antioxidant

589

compounds with the use of equivalent concentrations (EC1) defined as the concentration of

590

antioxidant with a reducing effect equivalent to 1 mmol/L Fe(II), and found that polyphenols

591

had lower EC1 values, and therefore higher reducing power, than ascorbic acid and Trolox.

592

Tannic acid and quercetin had the highest antioxidant capacity followed by gallic and caffeic

593

acids, while resveratrol showed the lowest reducing effect and carotenoids had no ferric

594

reducing ability. This inability of the FRAP method to assay thiols64 and carotenoids,45

595

possibly due to the kinetic inertness of high-spin Fe(III) and the problems associated with

596

mutual solubility of reagent and analyte in the same solvent medium, respectively, has also

597

been criticized in various research articles by other users of the method. Especially the

598

inability of the FRAP reagent to effectively oxidize biothiols make the method rather

599

ineffective in evaluting the TAC of intracellular fluids and human plasma/serum.11,64,65

600

Magalhaes et al.15 attribute this specific inadequacy of FRAP (i.e. an ET-reagent) to the mode

601

of action of thiols and carotenoids (i.e., antioxidants mainly acting by H-atom transfer). 25 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 26 of 105

602

However, if the problem had merely arisen from H-atom transfer, CUPRAC as another well-

603

known ET-based method would not have responded to carotenoids in aqueous acetone

604

solution.66 High-spin iron(III) in the reagent has half-filled d-orbitals responsible for kinetic

605

inertness, and thiol (RSH) oxidations usually proceed through thiyl (RS•) radical

606

intermediates which do not form appreciably at the acidic pH of the FRAP protocol.67

607

Another fact worthy of comment is that polyphenols with slow kinetic behaviors such as

608

caffeic acid, tannic acid, ferulic acid, p-coumaric acid, and quercetin cannot be fully oxidized

609

within the protocol time (i.e. typically 4 min) of the FRAP assay.14 FRAP reactions are

610

carried out in acidic medium in order to suppress Fe(III) hydrolysis (i.e. at pH 3.6) where

611

phenolic antioxidants are not dissociated, and therefore lower results than actual TAC are to

612

be expected because phenolates are oxidized much faster than the corresponding phenols.

613

FRAP and similar Fe(III)-reduction based ET assays were also criticized for producing Fe(II)

614

as the reduction product, which could give rise to the generation of reactive species (such as

615

hydroxyl radicals) upon Fenton-like reactions with H2O2, thereby causing ‘redox cycling’ of

616

phenolics and yielding erroneous TAC results.30 Antolovich et al.68 are of the opinion that the

617

measured ferric reducing capacity does not necessarily reflect antioxidant activity, but instead

618

provides a very useful ‘total’ antioxidant concentration, without measurement and summation

619

of the concentration of all antioxidants involved. Pulido et al.45 concluded that the antioxidant

620

efficiency of polyphenols depended on the extent of hydroxylation and conjugation.

621

Specifically for flavonoids, scientists investigating structure-activity relationships of

622

antioxidants suggested that the free radical scavenging ability increases when the following

623

criteria are met: (i) the presence of a 3’,4’-dihydroxy structure in the B ring; (ii) the presence

624

of a 2,3-double bond in conjunction with the 4-oxo group in the heterocycle, allowing for

625

conjugation between the A and B rings; (iii) the presence of 3- and 5-hydroxyl groups in the

626

A ring together with a 4-oxo function in the A and C rings (Figure 3).69,70 The results of

26 ACS Paragon Plus Environment

Page 27 of 105

Journal of Agricultural and Food Chemistry

627

Pulido et al.45 with flavonoids agreed with these criteria, e.g., quercetin, meeting all the listed

628

three conditions, was more potent than rutin (a flavonoid glycoside) and catechin (lacking

629

coplanarity due to the absence of 2,3-double bond, and therefore having hindered

630

conjugation/resonance stabilization over the whole molecule).

631 632 633 634 635

Figure 3

2.1.2.1.3. CUPRAC Assay

636 637

The CUPRAC assay of TAC determination is only 11-years old,40 but has branched into

638

various modified methods of antioxidant capacity/activity measurement associated with

639

Cu(II)-Cu(I) reduction in the presence of selective Cu(I)-stabilizing ligand, neocuproine (2,9-

640

dimethyl-1,10-phenanthroline). It has also been applied to various matrices containing both

641

hydrophilic and lipophilic antioxidants.

642

The main method is based on the absorbance measurement of the CUPRAC

643

chromophore, Cu(I)-neocuproine (Nc) chelate, formed as a result of the redox reaction of

644

antioxidants with the CUPRAC reagent, bis(neocuproine)copper(II) cation [Cu(II)-Nc], where

645

absorbance is recorded at the maximal light absorption wavelength of 450 nm (Figure 4).

646 647 648 649 650

Figure 4

651

with n-electron reductant antioxidants (AOX), according to Eq. 17:

The chromogenic oxidizing reagent of the developed CUPRAC method, Cu(II)-Nc, reacts

652 653 654 655 656

nCu(Nc)2 2+ + n-electron reductant (AOX) ↔ nCu(Nc)2+ + n-electron oxidized product + nH+ … (Eq. 17)

657

to the corresponding quinones (Ar=O), of ascorbic acid to dehdroascorbic acid, and of thiols

In this reaction, the oxidation of reactive Ar–OH groups of polyphenolic antioxidants

27 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 28 of 105

658

to the corresponding disulfides occur, while Cu(II)-Nc is reduced to the yellow-orange

659

colored Cu(Nc)2+ chelate. Although the concentration of Cu2+ ions is in stoichiometric excess

660

of that of Nc in the CUPRAC reagent for driving the redox equilibrium reaction to the right,

661

the actual oxidant is the Cu(Nc)22+ species and not the sole Cu2+, because the standard redox

662

potential of the Cu(II/I)-Nc is 0.6 V, much higher that of the Cu2+/Cu+ couple (0.17 V).71 The

663

reason is that Cu(I)-Nc is perfectly tetrahedral owing to the d10-electronic configuration of

664

Cu(I) having sp3 hybridization, while two molecules of neocuproine give a distorted

665

tetrahedral structure to Cu(II) having d9-configuration (i.e. known as Jahn-Teller effect in

666

coordination chemistry, which is enhanced when solvent molecules are also attached to

667

CuII(Nc)2 in octahedral coordination), thereby selectively stabilizing Cu(I) over Cu(II) (i.e. the

668

logarithmic stability constants of CuII(Nc)2 and CuI(Nc)2 are 12 and 19, respectively).72 As a

669

result, polyphenols are oxidized much more rapidly and efficiently with Cu(II)-Nc than with

670

Cu2+, and the amount of colored product [i.e., Cu(I)-Nc chelate] emerging at the end of the

671

redox reaction is equivalent to that of reacted Cu(II)-Nc.71 The liberated protons are buffered

672

in ammonium acetate medium, which provides a pH of 7.0 basically conforming to

673

physiological conditions. The highest antioxidant capacities in the CUPRAC method were

674

observed for epicatechin gallate, rosmarinic acid, epigallocatechin gallate, quercetin, fisetin,

675

epigallocatechin, catechin, caffeic acid, epicatechin, gallic acid, rutin, and chlorogenic acid in

676

this order,73,74 in accordance with theoretical expectations, because the number and position of

677

the hydroxyl groups as well as the degree of conjugation of the whole molecule are important

678

for easy electron transfer.70

679

Among the three substituted-phenanthrolines ‒used before in antioxidant or protein

680

assays‒ capable of selective stabilization of Cu(I) thereby increasing the Cu(II,I) reduction

681

potential, namely, Nc: neocuproine, BCS: 2,9-dimethyl-4,7-diphenyl-1,10-phenantroline

682

disulfonic acid,75 and bicinchoninic acid (BCA: 2-(4-carboxyquinolin-2-yl)quinoline-4-

28 ACS Paragon Plus Environment

Page 29 of 105

Journal of Agricultural and Food Chemistry

683

carboxylic acid),76 only neocuproine with the CUPRAC method has found wide use as an

684

effective tool in antioxidant research.40,41 Certainly, there are a number of reasons for this

685

choice due to the more diverse areas of usage of the cupric-neocuproine reagent. For example,

686

it is known that Cu(I)-BCS has a higher overall charge than Cu(I)-Nc due to the presence of

687

negatively-charged sulfonate groups on the phenanthroline ring, giving rise to stronger ion-

688

dipole interaction of the former with water molecules and subsequent cell membrane

689

impermeability. As a result, the Cu(I)-BCS method is expected to be less useful than

690

CUPRAC for the TAC assay of tissue homogenates. ET-based antioxidant assays may show

691

significant solvent dependencies and differences in proton-coupled electron transfer rate,77

692

and it has been shown by Çelik et al.78 that the cupric-BCS assay is not competent with

693

conventional CUPRAC using cupric-neocuproine reagent in regard to reaction kinetics and

694

response to lipophilic plasma antioxidants (e.g., β-carotene, α-tocopherol). The standard

695

reduction potential of the Cu(II,I)-BCS couple was reported to be Eo=0.844 V,79 a little higher

696

than those of most widely used ET-reagents, possibly affecting selectivity towards antioxidant

697

compounds. On the other hand, although BCA provides a longer wavelength (558 nm) than

698

Nc for cuprous chelate absorption which may seem advantageous at first glance for

699

preventing the possible interferences of plant pigments during measurement, Marques et al.80

700

recently discovered that in the BCA assay, the concentration of free Cu2+ ions cannot be

701

maintained in excess (i.e. required for the completion of certain oxidation reactions) because

702

of the precipitation of the complex.81 To guarantee a complete complexation without

703

precipitation, the BCA ligand must be added in stoichiometric ratio or in excess. Therefore,

704

the potential complexation of other metal ions that might be present in the sample (for

705

example Fe) cannot be ignored, and this situation limits the applicability of BCA.80

706

Owing to the favorable redox potential in neutral medium, the CUPRAC method has a

707

better chance of simulating physiologically important redox reactions of antioxidant

29 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 30 of 105

708

compounds, including serum antioxidants. In the normal CUPRAC method (CUPRACN), the

709

oxidation reactions of most food/biological antioxidants are essentially complete within 30

710

min. Flavonoid glycosides require acid hydrolysis to their corresponding aglycons for fully

711

exhibiting their antioxidant potency. Slow reacting antioxidants may need elevated

712

temperature incubation so as to complete their oxidation with the CUPRAC reagent.40,41

713

Although the protocol time period of the CUPRAC assay is set at 30 min, few antioxidants

714

with high redox potentials like naringin, naringenin and bilirubin may not reach absorbance

715

saturation so easily. On the other hand, since 80-90 % of the peak absorbances for a great

716

majority of antioxidants is reached within the first few minutes, online HPLC-post column

717

detection and voltammetric modifications of CUPRAC may allow a reaction time of 1 min for

718

antioxidants with the Cu(II)-neocuproine reagent.

719

The CUPRAC method of TAC assay has been successfully applied to antioxidants in

720

food plants, human serum, and to hydroxyl and superoxide radical scavengers. In the assay of

721

human serum antioxidants, hydrophilic antioxidants were measured after precipitation of

722

proteins with perchloric acid (trichloroacetic acid, ammonium sulfate and organic solvents are

723

other known protein-precipitation reagents), while lipophilic ones like α-tocopherol and β-

724

carotene were determined by n-hexane extraction, evaporation, followed by color

725

development in dichloromethane (DCM) of the Cu(I)-Nc charge-transfer complex formed

726

from their CUPRAC reaction.41 Since the CUPRAC chromophore, i.e. Cu(I)-neocuproine

727

cation, has a large molecular size, it has a very weak hydration sphere and therefore is easily

728

extracted into an organic solvent like DCM due to its low hydration energy (for divalent

729

chromophores

730

chromophore: phospho-tungsto-molybdate anion, this extraction is not so easy because of

731

enhanced ion-dipole interactions with solvent water molecules). This feature of the Cu(I)-Nc

732

chelate provides a great advantage for the CUPRAC method to be applicable to lipophilic

like

Fe(II)-tripyridyltriazine

cation

or

30 ACS Paragon Plus Environment

tetravalent

Folin-Ciocalteu

Page 31 of 105

Journal of Agricultural and Food Chemistry

733

antioxidants along with hydrophilic ones. In a miniaturized CUPRAC method without

734

preliminary separation of lipophilic and hydrophilic serum antioxidants, serum samples were

735

centrifuged after 10 % TCA precipitation, and CUPRAC was directly applied to the

736

supernate.82

737

Since essentially flavones and flavonols (and other flavonoids to a lesser extent) could

738

be chelated with lanthanum(III) in the form of basically nonpolar complexes, and ascorbic

739

acid (AA) either did not complex or formed very weak hydrophilic complexes under the same

740

conditions, AA assay with a high redox equilibrium constant of the CUPRAC reaction with

741

preliminary extraction of flavonoids as their La(III) complexes was possible.83 Although in

742

principle, ascorbate could be assayed by measuring the CUPRAC absorbance of a real

743

mixture before and after treating with the substrate-specific ascorbate oxidase enzyme, many

744

constituents in plant extracts may inhibit this enzyme causing erroneous results. Lipophilic

745

and hydrophilic antioxidants (e.g., β-carotene, α-tocopherol, AA, quercetin, etc.) could be

746

simultaneously assayed with a modified CUPRAC method in the same solvent medium of

747

acetone- water (9:1, v/v) with the aid of their inclusion complexes formed with 2 % methyl-β-

748

cyclodextrin (M-β-CD), because this oligosaccharide can form inclusion complexes (Figure

749

5) with lipophilic antioxidants in the interior part while the outer part binds hydrophilic

750

antioxidants.84

751 752 753 754

Figure 5 The CUPRAC assay could be further modified to fit the needs of scavenging activity

755

determinations of reactive oxygen and nitrogen species (ROS/RNS) such as hydrogen

756

peroxide, superoxide anion and hydroxyl radicals, where either the original probe or the

757

converted product had CUPRAC reactivity. In measuring the hydroxyl radical scavenging

758

activity of certain water-soluble compounds (metabisulfite, thiourea, glucose, lysine, etc.), the

759

probes of p-aminobenzoate, 2,4-dimethoxy-benzoate, and 3,5-dimethoxybenzoate were used 31 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 32 of 105

760

to detect hydroxyl radicals, and the •OH scavenging rate constants of these compounds were

761

determined by competition kinetics.85 In the measurement of hydroxyl radical scavenging

762

activities of polyphenolics, special measures were taken so as to prevent the redox cycling of

763

phenolic compounds that could otherwise produce unrealistic results. For this purpose, the

764

Fenton reaction was stopped at the end of the 10th minute with the addition of catalase to

765

annihilate hydrogen peroxide and cease •OH production, and the dihydroxybenzoates formed

766

from the salicylate probe under hydroxyl radical attack (Figures 6a and 6b) were measured

767

with the CUPRAC method, rate constants being calculated with competition kinetics.12

768 769 770 771 772

Figure 6

773

with xanthine–xanthine oxidase (X–XO), and the inhibition of the enzyme was measured

774

upon addition of polyphenolics to the system (Figure 7).86

775 776 777 778 779 780 781 782

In another modified CUPRAC method, the superoxide anion radical was generated

Figure 7. Xanthine oxidase (XO) inhibitors obstruct the formation of CUPRAC-reactive uric acid from xanthine, thereby enabling a modified CUPRAC assay for measuring XO-inhibiting antioxidant activity.

The hydrogen peroxide scavenging (HPS) activity of the polyphenolics was measured

783

in the presence of Cu(II) (as catalyst) with the HPS-CUPRAC method.87 A low-cost optical

784

antioxidant sensor (CUPRAC sensor) was developed by immobilizing the Cu(II)-Nc reagent

785

onto a perfluorosulfonate cation-exchange polymer membrane matrix (Nafion®),73 and the

786

colored Cu(I)-Nc cation was produced on the membrane without diffusing into solution. This

787

membrane sensor provided great ease and convenience to TAC determinations, like a

788

sensitive pH paper immersed in solution for hydrogen ion activity determination (Figure 8).

789 790 791

Figure 8. The cationic chelate of the CUPRAC reagent, Cu(Nc)22+, is electrostatically held on the sulfonate groups of a cation-exchange membrane (Nafion), producing a CUPRAC 32 ACS Paragon Plus Environment

Page 33 of 105

Journal of Agricultural and Food Chemistry

792 793 794 795

antioxidant sensor; antioxidants reduce this membrane-held chelate to the cuprous chromophore, Cu(Nc)2+, showing maximal absorption at 450 nm.

796

determination of polyphenols in complex plant matrices. This method combines

797

chromatographic separation, constituent analysis, and post-column identification of

798

antioxidants (Figure 9) in plant extracts. Antioxidant polyphenols in complex samples can be

799

separated on a C18 HPLC column (diode-array detected at 280 nm) and further react with the

800

Cu(II)-Nc reagent in a post-column reactor to yield the Cu(I)-Nc chromophore detected at 450

801

nm. Thus, twice as much information can be extracted from the same sample using these two

802

chromatograms, i.e. their separability through a C18 column and their CUPRAC reactivity in

803

the post-column. This robust online chromatographic method enables individual

804

detection/quantitation of antioxidant constituents as well as total measurement of TAC;

805

moreover, non-antioxidants are not detected in the post-column chromatogram, increasing

806

selectivity of the method.88

807 808 809 810 811 812 813 814

Figure 9. The chromatogram of green tea extract showing conventional HPLC (with 280-nm detection: positive trace chromatogram) and on-line post-column HPLC-CUPRAC assay (with 450-nm detection: negative-trace chromatogram); notice that gallic acid (GA) and tea catechins (CT, EC, EGC, ECG, EGCG) are identified in both chromatograms, while the CUPRAC non-reactive caffeine, symbolized as (C), gave a peak only in the upper chromatogram but not in the lower one, because it is not an antioxidant compound.

815

ammonium acetate, should be replaced with urea at the same pH in order to prevent re-

816

precipitation of dissolved proteins. CUPRAC in urea buffer also responded to thiol-containing

817

proteins in food and serum.11,64 Another modified CUPRAC method is comprised of a tert-

818

butylhydroquinone (TBHQ) probe with the phenazine methosulphate/β-nicotinamide adenine

819

dinucleotide (PMS/NADH) non-enzymic O2•– generating system for superoxide radical

820

scavenging activity (SRSA) assay of thiol-type antioxidants (e.g., GSH, cysteine), amino

821

acids (e.g., serine, threonine), plasma antioxidants (e.g., bilirubin, albumin), and other

A novel online HPLC-CUPRAC method was developed for the selective

In protein TAC determination, the classical buffer of the CUPRAC reaction, i.e.

33 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 34 of 105

822

antioxidants (e.g., methionine); the SRSA method is based on the measurement of the

823

CUPRAC absorbance of the remaining TBHQ in the reaction medium (TBHQ is CUPRAC-

824

reactive while its oxidation product is not (Figure 7), and this probe is isolated by ethyl

825

acetate extraction from other CUPRAC-reactive interferents remaining in the aqueous

826

phase).89

827 828

Figure 7

829 830

The CUPRAC assay together with its modifications for ROS scavenging

831

measurements have been summarized, and the methodology has been demonstrated to have

832

certain advantages over other similar ET-based TAC methods, mentioned in a comprehensive

833

review by Özyürek et al.:8

834

(i) The CUPRAC reagent, being an outer-sphere electron-transfer agent, is capable of rapidly

835

oxidizing thiol-type antioxidants, whereas iron(III)-based ET-methods like FRAP may only

836

measure limited thiols like GSH with serious negative error, possibly due to the kinetic

837

inertness of high-spin Fe(III) and to the inadequate formation of thiyl radicals (i.e.

838

intermediary species of thiol oxidation) in acidic medium. Since the redox potential of

839

oxidized and reduced forms of glutathione (EoGSSG/2GSH) is the basic indicator of the biological

840

conditions of a cell, and GSH acts as reconstituent of intercellular ascorbic acid from the

841

dehydroascorbic acid, an ET assay should be capable of accurately measuring glutathione

842

among plasma antioxidants.

843

(ii) The redox potential of Cu(II,I) chelated to necuproine is only 0.6 V, close to that of

844

ABTS•+ /ABTS (Eo=0.68 V) and FRAP (Eo=0.70 V), all versus NHE. Simple sugars and citric

845

acid –which are not classified as antioxidants– are not oxidized with the CUPRAC reagent.

34 ACS Paragon Plus Environment

Page 35 of 105

Journal of Agricultural and Food Chemistry

846

On the other hand, the ferric/ferrous potential in the presence of ortho-phenanthroline or

847

batho-phenanthroline-type ligands is much higher, adversely affecting selectivity.

848

(iii) The reagent is much more stable and readily accessible than the chromogenic radical

849

reagents

850

dihydrochloride (DMPD)).

851

(iv) The CUPRAC method shows versatility to the determination of both hydrophilic and

852

lipophilic antioxidants, because the CUPRAC chromophore, bis(neocuproine)copper(I)

853

chelate, has unipositive charge having less ion-dipole interaction with water, and the chelate

854

rings are essentially hydrophobic. Thus it is compatible with aqueous and organic solvents

855

(alcohols, acetone, dichloromethane, etc.) and alcohol-water mixtures. The robustness of the

856

CUPRAC assay to different solvents has been recently confirmed by Christodouleas et al.90 in

857

the TAC assay development study for edible oils.

858

(v) The redox reaction giving rise to the Cu(I)-Nc chromophore is relatively insensitive to a

859

number of parameters (e.g., air, sunlight, humidity, and to a certain extent pH) adversely

860

affecting radical reagents such as DPPH.

861

(vi) The CUPRAC reagent can be adsorbed on a perfluorosulfonate cation-exchanger

862

membrane enabling the manufacture of a low-cost, linear-response antioxidant sensor.

863

CUPRAC is also adaptable to online-HPLC applications with the use of a post-column

864

reactor, enabling sensitive determination of antioxidants individually.

865

(vii) CUPRAC usually gives perfectly linear absorbance/concentration curves (r ≈ 0.999) over

866

a wide concentration range, as opposed to certain other methods yielding polynomial curves.

867

The molar absorptivity (extinction coefficient) for n-electron reductants, i.e. (7.5–9.5x103 n)

868

M-1cm-1, is sufficiently high to enable sensitive determination of most phenolic antioxidants.

869

(viii) The CUPRAC spectrophotometric method obeys Beer’s law in regard to the additivity

870

of absorbances due to individual antioxidant constituents, because a single chromophore,

(e.g.,

ABTS,

DPPH,

galvinoxyl

and

N,N-Dimethyl-p-phenylenediamine

35 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 36 of 105

871

cuprous-neocuproine, is formed upon reduction of the CUPRAC reagent with antioxidants.

872

Consequently, the CUPRAC-TAC values of antioxidants in complex mixtures are perfectly

873

additive (e.g., the TAC of a phenolic mixture is equal to the sum of individual antioxidant

874

capacities of its constituent polyphenols).

875

(ix) CUPRAC operates at nearly physiological pH (pH 7 of ammonium-acetate buffer) as

876

opposed to the unrealistic acidic conditions (pH 3.6) of FRAP or alkaline conditions (pH 10)

877

necessary for phenols to dissociate protons in the Folin-Ciocalteu assay. At more acidic

878

conditions than the physiological pH, the reducing capacity may be suppressed due to

879

protonation on antioxidant compounds, whereas in more basic conditions, deprotonation of

880

phenolics enhances a sample’s reducing capacity, thereby causing unrealistic TAC

881

measurements.

882

(x) Since the Cu(I) ion emerging as a product of the CUPRAC redox reaction is in a

883

coordinatively saturated state (i.e. two molecules of neocuproine tetrahedrally coordinate the

884

cuprous ion), it cannot act as a prooxidant that may cause oxidative damage to biological

885

macromolecules in body fluids. Fe(III)-based assays were criticized for producing Fe2+, which

886

may act as a prooxidant to produce •OH radicals as a result of its reaction with H2O2, and

887

subsequently cause a ‘redox cycling’ of antioxidants during the assay, yielding unreliable

888

results. Ferric or ferrous iron, even in full octahedrally-coordinated (e.g., EDTA-chelated)

889

state, can catalyze the decomposition of hydrogen peroxide to reactive species.91 On the other

890

hand, it was experimentally shown that the stable Cu(I)-Nc chelate did not react with H2O2,

891

but the reverse reaction (i.e. oxidation of H2O2 with Cu(II)-Nc) is possible, excluding the

892

possibility of redox cycling of antioxidants with the Cu(I)-Nc product.

893

(xi) The CUPRAC method has recently been implemented in microplate and flow modes,

894

allowing the in vitro assessment of antioxidant capacity of endogenous and dietary molecules

895

as well as the TAC determination of human biological samples.92

36 ACS Paragon Plus Environment

Page 37 of 105

Journal of Agricultural and Food Chemistry

896

(xii) As noteworthy experiences of CUPRAC users, Gorinstein research groups stated that, as

897

an advantage over other electron-transfer-based assays, the CUPRAC test gave reproducible

898

values that were acceptable in regard to its realistic pH close to physiological pH in various

899

food extracts (garlic,93 onion, kiwi, etc.). Bean et al.65 made a comparative evaluation of

900

antioxidant reactivity within obstructed (i.e. resulting from the attack of ROS/RNS in cyclic

901

ischemia and reperfusion) and control rabbit urinary bladder tissue using FRAP and CUPRAC

902

assays, and found that that CUPRAC, but not FRAP, could detect a significant decrease in the

903

reactivity of antioxidants found within the obstructed bladder tissue as compared to the

904

control bladder tissue in both the muscle and mucosa. Bean et al.65 concluded that, as the

905

CUPRAC assay was responsive to hydrophilic, lipophilic, and thiol-containing antioxidants at

906

physiological pH, it was a much better tool to analyze the reactivity found within tissues.

907 908 909

2.1.2.1.4. Ferricyanide (Hexacyanoferrate(III))-Prussian Blue Assay

910 911

The ferricyanide-Prussian blue assay is based on the following chemical reactions:

912 913

Fe(CN)63− + ArOH → Fe(CN)64− + ArO• + H+ … (Eq. 18)

914

Fe(CN)64− + Fe3+ + K+ → KFe[Fe(CN)6] … (Eq. 19) with λmax=700 nm.

915 916

In the conventional method,46 the hexacyanoferrate(III) (common name: ferricyanide) reagent

917

is first incubated in (H2PO4-/HPO42-) buffer at pH 6.6 with antioxidants (at 50°C for 20 min),

918

and the reduction product, hexacyanoferrate(II) (common name: ferrocyanide), combines with

919

the later added Fe(III) to produce Prussian blue, suspended in the medium. The method is also

920

referred to as the ‘reducing power’ assay.94

37 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 38 of 105

921

Although the Fe(III,II) standard reduction potential is 0.77 V causing nonspecific

922

oxidation of any species having a redox potential smaller than this value,59 suitable selection

923

of ligands may bring this value close to the range of common food and biological antioxidants

924

(i.e., having standard potentials in the range of 0.2–0.6 V). In the case of cyanide complexes

925

of iron in hexacyanoferrate complex ions, the logarithmic stability constant of Fe(III)

926

complex is greater than that for Fe(II) (i.e. Log β6 for Fe(CN)63− and Fe(CN)64− are 31 and 24,

927

respectively). Therefore, according to the Nernst equation, the reduction potential for

928

hexanocyanoferrate (III,II) couple is 0.36 V versus NHE, less than that of Fe(III,II). This

929

potential may not be sufficient for oxidizing certain antioxidants having a reduction potential

930

(of the ArO•/ArOH couple) Eo > 0.4 V, and therefore modification of the conventional

931

ferricyanide assay46 was a requirement for comprising diverse antioxidants. The wavelength

932

of maximum absorption shifts to longer wavelengths in the order of ferric-complexing

933

ligands: ortho-phen < batho-phen < tripyridyltriazine (FRAP) < ferricyanide. The

934

bathochromic shift in λmax and band broadening was strongest for ‘Prussian blue’ in the

935

ferricyanide method, because the bonding and antibonding energy levels were closest in

936

Fe[Fe(CN)6]− due to the interchanging oxidation states of iron centers in this complex salt.

937

Longer wavelengths almost always constitute an important advantage in spectrophotometric

938

method selection, because most plant pigments as well as some antioxidants show significant

939

absorption at shorter wavelengths of the visible region, close to the UV range of the visible

940

spectrum.48

941

The conventional ferricyanide assay was first modified by incorporating Fe(III) to the

942

ferricyanide reagent in acidic medium and incubating at elevated temperature (30 min

943

incubation at 50°C), but gave rise to overoxidation of certain antioxidants (especially of

944

hydroxycinnamic acids like caffeic and ferulic acids) causing distinct deviations from

945

linearity of calibration curves.48 Then the authors introduced a second modification of the

38 ACS Paragon Plus Environment

Page 39 of 105

Journal of Agricultural and Food Chemistry

946

method, by adding Fe(III) at the start and optimizing the acidity of the incubation medium as

947

pH=1.6 (so as to prevent Fe(III) hydrolysis), and by adding the anionic surfactant SDS to

948

stabilize the negatively-charged Prussian blue complex ion: Fe[Fe(CN)6]-.47 Compared to the

949

original method,46 stabilization with SDS enabled a color development time of 30 min at

950

room temperature (instead of the 2-min reaction time of Oyaizu to avoid Prussian blue

951

precipitation) and a wavelength (λmax) shift from 700 nm to 750 nm. The modified procedure

952

was named as the ‘ferric-ferricyanide assay’, because it was not clear whether Fe(III) or

953

ferricyanide was the actual oxidant. Consequently, the oxidation equilibria for antioxidants by

954

the combined (Fe(III) + Fe(CN)63−) reagent significantly shifted to the right, possibly

955

increasing the potential above that of Fe(III,II) couple, owing to the formation of insoluble

956

Prussian blue, i.e. if the actual oxidant was Fe(III), then its reduced form, Fe(II), would again

957

produce the same Prussian blue product with the ferricyanide constituent of the reagent:

958 959 960 961 962 963

Fe3+ + ArOH → Fe2+ + ArO• + H+ … (Eq. 20) Fe2+ + Fe(CN)63− + K+ → KFe[Fe(CN)6] … (Eq. 21) with λmax=750 nm. Finally, with the third modification of the method, Berker et al.95 were able to measure

964

lipophilic (e.g., α-tocopherol, BHT, and β-carotene) and hydrophilic antioxidants in the same

965

solution comprising 1:9 (v/v) H2O–acetone with or without 2% methyl-β-cyclodextrin; this

966

was necessary, as the original ferricyanide method could only assay hydrophilic antioxidants.

967

This modified assay was not adversely affected from citric acid and simple sugars, and proved

968

to be additive for TAC values of complex mixtures.95

969 970

2.1.2.1.5. ET-based Spectrophotometric Assays Involving Strongly Oxidizing Reagents

971

(Ce(IV), Cr(VI), and Mn(VII) Assays)

972

39 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 40 of 105

973

Strong oxidizing agents such as Ce(IV), Cr(VI) and Mn(VII) may be used as chromogenic

974

TAC reagents only if their oxidizing power are decreased to the level of specifically oxidizing

975

common antioxidants but not other organic substances (i.e. their redox potentials should be

976

brought to roughly 0.6−0.7 V of widely used TAC reagents). This can usually be

977

accomplished by increasing the working pH (e.g., the extent of chromate(VI) and

978

permanganate(VII) oxidations is a function of H+-ion concentration) and/or by selectively

979

stabilizing the higher oxidation state of the redox couple (e.g., by preferential complexation of

980

Ce(IV) over Ce(III) with sulfate) so as to decrease the Nernst potential (E = Eo + (RT/nF) Ln

981

[cox/cred]), where E and Eo are the instantaneous and standard values of the reduction potential,

982

respectively, R: universal gas constant, T: absolute temperature, n: number of electrons

983

involved in half-cell reaction, F: Faraday’s constant, and cox and cred are the concentrations of

984

the oxidized and reduced forms, respectively, of the redox-active constituent of the TAC

985

reagent). Another disadvantage of the use of strong oxidizing agents in TAC assays is their

986

hydrophilicity, i.e. they cannot be applied to lipophilic antioxidant testing.

987

Özyurt et al.51 developed a simple, sensitive, and low-cost indirect spectrophotometric

988

method for the determination of Ce(IV) reducing antioxidant capacity (CERAC) of plant

989

extracts, based on the oxidation of antioxidants with cerium(IV) sulfate in dilute sulfuric acid

990

at room temperature. The Ce(IV) reducing antioxidant capacity (CERAC) of the sample was

991

measured under controlled conditions of oxidant concentration and pH such that antioxidants

992

but not other organic compounds would be oxidized. The spectrophotometric determination of

993

the remaining Ce(IV) at 320 nm was performed after all antioxidants in solution were

994

oxidized. Blank correction of significantly absorbing plant extracts at 320 nm could be made

995

with the aid of spectrophotometric titration. The trolox equivalent antioxidant capacities

996

(TEAC coefficients) of the tested antioxidant compounds were correlated to those found by

997

ABTS and CUPRAC methods. Since the TEAC coefficients (found by CERAC) of naringin–

40 ACS Paragon Plus Environment

Page 41 of 105

Journal of Agricultural and Food Chemistry

998

naringenin and rutin–catechin pairs were close to each other, this assay was advantageous to

999

accomplish the simultaneous hydrolysis of flavonoid glycosides to the corresponding

1000

aglycones and their subsequent oxidation such that the hydrolysis products exhibed

1001

antioxidant capacities roughly proportional the number of –OH groups in a phenolic

1002

molecule. Özyurt et al.52 further developed the CERAC method by employing a medium of

1003

(0.3 M H2SO4 + 0.7 M Na2SO4) to finely tune the Nernst potential of the Ce(IV)/Ce(III)

1004

couple by maintaining sufficient acidity while selectively complexing Ce(IV) with sulfate in

1005

preference to Ce(III) so as to oxidize true antioxidants but not citric acid or simple sugars.

1006

The TEAC coefficients of this modified CERAC procedure for antioxidants were in the order

1007

of quercetin > rutin > gallic acid > catechin > caffeic acid ≥ ferulic acid > naringenin ≥

1008

naringin > trolox ≥ ascorbic acid, in accordance with those found by other antioxidant assays.

1009

It was also possible for Özyurt et al.53 to measure the fluorescence of the reduction product,

1010

Ce(III), with excitation at 256 nm and emission at 360 nm, and then correlate this

1011

fluorescence intensity to the TAC value of a sample. By measuring the produced Ce(III)

1012

fluorometrically instead of the remaining Ce(IV) spectrophotometrically, the linear range was

1013

widened (e.g., 5.0 × 10−7–1.0 × 10−5 M for quercetin) and the possible interferences of plant

1014

pigments for absorbance measurement at 320 nm were eliminated (however, the linear

1015

correlation coefficient in fluorescence was lower than in spectrophotometry due to the

1016

fluorescence-quenching effect of Ce(IV)).

1017

Işık et al.50 developed the Cr(VI) reducing antioxidant capacity (CHROMAC) assay,

1018

involving the reduction of chromate(VI) with antioxidants to Cr(III) in acidic solution at pH

1019

2.8, and after 50-min reaction time, the remaining Cr(VI) was spectrophotometrically

1020

measured with 1,5-diphenylcarbazide (DPC) at 540 nm. The in situ-formed Cr(III) complex

1021

with the oxidation product of DPC (i.e. diphenylcarbazone) indicated the remaining chromate,

1022

and Cr(VI) consumption was correlated to antioxidant concentration in the sample. The

41 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 42 of 105

1023

authors found comparable results to ABTS and CUPRAC for a number of plant extracts.

1024

However, the TAC order of common antioxidants in CHROMAC did not well agree with

1025

those of other ET-based assays (e.g., rosmarinic acid and quercetin were found to be less

1026

potent than ascorbic acid, based on the comparison of TEAC values). If no measures were

1027

taken for regulating the Nernst potential of Cr(VI)/Cr(III) redox couple, as in chemical

1028

oxygen demand (COD) tests performed in water treatment, the acidic dichromate reagent

1029

would oxidize all the organic load of solution without discriminating antioxidant compounds.

1030

Potassium permanganate is a strong oxidant widely used in analytical chemistry for

1031

redox titrations without an external indicator because of the intrinsic violet color of the

1032

reagent. A critical and comprehensive review of acidic potassium permanganate

1033

chemiluminescence was presented, together with a discussion on reaction conditions, the

1034

relationship between analyte structure and chemiluminescence intensity, and its application to

1035

determine a variety of compounds including antioxidants.96 Francis et al.97 reported the use of

1036

acidic potassium permanganate as a chemiluminescence reagent to rapidly assess the

1037

antioxidant status of fruit juices, teas and other beverages. In the acidic KMnO4

1038

spectrophotometric assay of reducing capacity for antioxidants,98 the sample was oxidized

1039

with acidic permanganate leading to sample discoloration until no color was observed;

1040

subsequent decrease of potassium permanganate concentration was determined with the use

1041

of a calibration curve of absorbance at 535 nm versus concentration.99 In the original assay,

1042

Cacig et al.98 also observed MnO2 particles in suspension, showing the non-stoichiometric

1043

character of oxidation (instead of a neat Mn(VII)-Mn(II) reduction). Although the results of

1044

this method were claimed to correlate with those of other reducing assays and acidic

1045

permanganate was assumed to oxidize phenol by forming phenoxyl-radical and manganic

1046

acid (H2MnO4) in the slowest step of a series of oxidation reactions, it is obvious that

42 ACS Paragon Plus Environment

Page 43 of 105

Journal of Agricultural and Food Chemistry

1047

permanganate in sulfuric acid medium would non-specifically oxidize any organic substance

1048

and the measured parameter would be ‘total organic status’ of a sample rather than its TAC.

1049 1050

2.1.2.2. Electrochemical Methods

1051 1052

Direct electrochemical sensing methods for in vitro antioxidant capacity assessment have

1053

been reviewed by Blasco et al.100 Cyclic voltammetry (CV) is an electrochemical technique

1054

in which a sample is introduced to a reaction chamber with three electrodes: working

1055

electrode (such as glassy carbon (GC)), reference electrode (e.g., Ag/AgCl), and auxiliary

1056

electrode (e.g., Pt wire), where an increasing potential is applied to the working electrode and

1057

current intensity versus potential is recorded.33 Although not all antioxidants can give their

1058

electrons to the GC electrode at an appreciable rate, most antioxidants are CV-active reducing

1059

agents and can therefore be assayed by CV on the basis of their redox potentials. For example,

1060

in cyclic voltammetry utilizing a GC electrode, chlorogenic and caffeic acids showed well-

1061

defined reversible waves in weakly acidic solution, while their electrooxidation assumed a

1062

quasi-reversible character at higher pH due to possible polymerization reactions at the

1063

electrode surface.101 Chevion et al.102 made some pioneering studies on the electrochemical

1064

determination of antioxidant capacity, and defined cyclic voltammetry as a simple, sensitive

1065

and reliable method for determining the TAC of human plasma originating from low

1066

molecular-weight antioxidants (LMWA). Half-wave potential (E1/2) indicated the specific

1067

constituent of the LMWA and its ability to donate electron(s), whereas current intensity at

1068

half-wave potential (Ia) indicated the concentration of this constituent. The E1/2 and Ia values

1069

reflected the antioxidant capacity of the plasma, while the change of Ia upon exposure to

1070

copper ions, ionizing radiation and peroxyl radicals represented the severity of the oxidative

1071

stress induced. In their later work,103 the authors suggested another parameter better

43 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 44 of 105

1072

correlating with antioxidant concentration, namely the AUC around the anodic wave

1073

potential: E1/2. Using the AUC approach, LMWA constituents of human plasma and animal

1074

tissues were identified and further validated by reconstruction of the CV tracing and by

1075

HPLC-electrochemical detection. As the changes for individual constituents within a single

1076

AC wave could be different, the changes in AUC would better represent the residual

1077

antioxidant capacity and allow better quantitation of the loss in a specific component upon

1078

oxidative stress. Chevion et al.102 identified two critical E1/2 values for human plasma, namely

1079

420 ± 25 mV (mainly derived from ascorbate and urate constituents of LMWA) and 920 ± 25

1080

mV (derived from unidentified constituents, excluding simple and protein thiols). Kohen et al.

1081

later showed that this group of mostly unidentified LMWA giving rise to the high-potential

1082

anodic wave could be related to the oxidized form of lipoic acid.104 Spiking brain tissue

1083

samples with relatively high concentrations (0.5 mM) of other antioxidants (carnosine,

1084

tryptophan, and melatonin) yielded an increase in the amplitude of the second anodic wave.103

1085

The authors also found that when the wave was comprised of several constituents

1086

characterized by close but different E1/2 values, a change in the relative concentrations of the

1087

constituents would cause a small shift in the E1/2 of their combined anodic wave.102 Plasma

1088

samples should be preserved with heparin when necessary, but not with EDTA which gave a

1089

specific anodic wave at E1/2 > 900 mV. The major disadvantages of CV are low sensitivity for

1090

antioxidants (usually in several micromolar range) and the necessity for frequent electrode

1091

cleansing before each measurement due to adsorption of proteins and other macromolecules

1092

from biological fluids onto the glassy carbon working electrode.33

1093

Piljac-Žegarac et al.105 used CV to study the electrochemical properties of antioxidants

1094

in fruit tea infusions as well as to estimate the antioxidant capacity. The easiest oxidized

1095

compound (at approximately 130 mV) was ascribed to the oxidation of the ene-diol of

1096

ascorbic acid. A pronounced anodic current peak (corresponding to a quasi-reversible redox

44 ACS Paragon Plus Environment

Page 45 of 105

Journal of Agricultural and Food Chemistry

1097

process, reflecting oxidation to a stable quinone) observed at 440 mV in all analyzed fruit teas

1098

indicated that ortho-dihydroxy-phenol and gallate groups were the major contributors to the

1099

antioxidant capacity of investigated teas. The less oxidizable compounds presented an

1100

irreversible oxidation process between 670 and 700 mV, which was ascribed to the oxidation

1101

of the monophenol group or the meta-diphenols on the A-ring of flavonoids that led to a

1102

phenoxy radical or a phenoxonium ion undergoing successive secondary reactions. The

1103

antioxidant capacity of these fruit teas was determined by estimating the integrated area

1104

(AUC) under the peak up to 600 mV.

1105

Electrochemical techniques of antioxidant characterization consist of CV, differential

1106

pulse (DPV) and square-wave voltammetry (SWV), proposed as useful tools to investigate the

1107

electrochemical behavior of phenolic compounds in different food samples in conjunction

1108

with carbon, diamond and graphite electrodes.106,107 DPV is one of the most sensitive

1109

techniques, and has a received a great deal of attention in recent years.

1110

CV methods have also been described to detect ascorbic acid, citric acid, and sugars in

1111

both food products and pharmaceuticals and to consider their influence on the oxygen

1112

reduction process, but this approach is restricted due to the electrochemically inactive nature

1113

of these compounds in the potential range of O2 reduction.108 Qualitative assessment of wine

1114

phenolics based on reducing strength was also realized by using CV at a GC electrode;

1115

although in this approach, the AUC, namely the charge passed to 0.5 V potential during CV

1116

recording, is understood as a better estimate of the concentration of polyphenols with low

1117

oxidation potentials, quantitation efforts of all reducing compounds may only provide a

1118

qualitative picture in a complex mixture such as wine mainly because the magnitude of the

1119

response is not identical on a molar basis for dissimilar compounds.109 The antioxidant

1120

capacities of several drugs containing acetylsalicylic acid were evaluated using a SOD-based

1121

biosensor method in comparison to ORAC-fluorometric and DPV methods, but although the

45 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 46 of 105

1122

DPV-based method generally seemed to be sufficiently in line with the results of the other

1123

two methods, its precision was rather poor (RSD > 10%).110 Novak et al. used square-wave

1124

voltammetry for investigating the electrochemical behavior of major green tea compounds

1125

such as epigallocatechin gallate, epigallocatechin and gallic acid.106 Magarelli et al. developed

1126

and validated a sensitive DPV method using a GC electrode for the determination of total

1127

phenolic acids in cotton cultivars;107 however, the authors studied only four polyphenolics

1128

(i.e. caffeic, chlorogenic, gallic and gentisic acids) and presented one anodic peak at

1129

approximately 0.4 V that was assigned to the oxidation of phenolic hydroxyls leading to the

1130

formation of o-quinone via semiquinone form, and therefore, it may be argued whether these

1131

four easily-oxidized phenolic acids are a true representative of ‘total phenolic acids’ having

1132

diverse oxidation potentials.

1133

There is very limited study about the usage of GC electrodes as electrochemical TAC

1134

sensors. Milardović et al.111 developed an amperometric method, based on the reduction of

1135

DPPH• at the GC electrode, for measuring the antioxidant activity of pure compounds and

1136

their real mixtures such as tea, wine and other beverages; since the cyclic voltammograms of

1137

a number of water- and ethanol-soluble common antioxidants gave either irreversible or

1138

undefined oxidation peaks, electrochemical reduction of DPPH• was conducted in the

1139

presence and absence of antioxidants, but potential selection was critical due to the

1140

electrochemical interferences of caffeic acid and trolox. Milardović et al.111 further concluded

1141

that amperometric and spectrophotometric DPPH•-based TAC methods had similarities, since

1142

both methods are based on the same reaction thermodynamics and kinetics. The

1143

electrochemical ABTS•+ assay is based on measuring catalytic voltammetric currents caused

1144

by antioxidants acting as reductant toward the electrochemically generated ABTS 2+ (thereby

1145

enabling reoxidation of ABTS•+ on the electrode) in edible oils; this oxidative voltammetric

1146

current intensity increased with an increase in antioxidant concentration, enabling TAC

46 ACS Paragon Plus Environment

Page 47 of 105

Journal of Agricultural and Food Chemistry

1147

determination of edible oils. This method produced rather high blank values in the Trolox

1148

calibration curve, and Trolox addition to sunflower oil matrices did not yield perfectly linear

1149

responses.112 In an attempt to adapt the Ce(IV)-reducing TAC assay51 to electroanalytical

1150

chemistry, Ferreira and Avaca113 measured the ability of eight different compounds in

1151

reducing Ce4+ by chronoamperometric measurement of the remaining Ce3+ species, and found

1152

the TAC order as: tannic acid >> quercetin > rutin > gallic acid ≈ catechin > ascorbic acid >

1153

BHA > Trolox, agreeing well with FRAP. The electrochemical behavior of the Cu(Nc)22+

1154

complex was recently studied by cyclic voltammetry at a GC electrode.114 The

1155

electroanalytical method was based on the reduction of Cu(Nc)22+ to Cu(Nc)2+ by antioxidants

1156

and electrochemical detection of the remaining Cu(II)–Nc (unreacted complex), the difference

1157

being correlated to antioxidant capacity of the analytes. The calibration curves of individual

1158

compounds comprising polyphenolics and vitamin C were constructed, and their response

1159

sensitivities and linear concentration ranges were determined. The reagent on the GC

1160

electrode retained its reactivity toward antioxidants, and the measured TEAC values of

1161

various antioxidants suggested that the reactivity of the Cu(II)–Nc reagent is comparable to

1162

that of the solution-based spectrophotometric CUPRAC assay. This electroanalytical method

1163

better tolerated sample turbidity and provided higher sensitivity (i.e. lower detection limits) in

1164

antioxidant determination than the spectrophotometric assay.114

1165

In a review work, Prieto-Simon et al.115 made a generalization that electroanalytical

1166

biosensor-originated antioxidant activity/capacity measurements based on the reduction of

1167

hazard caused by O2•− essentially involve the use of (i) Cyt c heme protein, (ii) SOD enzyme,

1168

and (iii) DNA. Superoxide anion determination using a Cyt c-based sensor was reported not

1169

to be so selective mainly because this heme protein is not specific for O2•− and can

1170

simultaneously reduce endogeneous H2O2 in biological systems, whereas SOD-based

1171

biosensors were claimed to be more selective and sensitive.115 When using DNA-based

47 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 48 of 105

1172

bioelectrosensors, the oxidizibility of DNA bases (i.e. maintaining DNA integrity) in the

1173

absence and presence of antioxidants was taken as a measure of antioxidant activity.

1174

(i) O2•− was produced by the xanthine-XOD enzyme system, using a Cyt c-modified

1175

electrode, where Cyt c was reported to communicate with the nano-Au electrode through self-

1176

assembled monolayers of short chain alkanethiols. The immobilised Cyt c was reduced by

1177

O2•− and rapidly regenerated at the surface of the electrode polarized at the oxidation

1178

potential, where the current generated by electron transfer from the radical to the electrode

1179

through Cyt c was proportional to the radical concentration.116 In this process, antioxidants,

1180

when present, quenched the radicals and decreased their concentration reflected in a decrease

1181

of oxidation current. Using this bioelectrosensor, an antioxidant activity sequence was

1182

established for flavonoids in decreasing order: flavanols > flavonols > flavones > flavonones

1183

> isoflavonones.117

1184

(ii)

1185

monolayers) Au nanoparticles-coated carbon fiber microelectrodes, enabling direct electron

1186

transfer with O2•−,118 catalyzing their dismutation to O2 and H2O2 :

1187 1188 1189 1190 1191 1192

SOD enzyme was immobilized onto cysteine-functionalized (via self-assembled

SOD (Cu2+) + O2•− → SOD (Cu+) + O2 … (Eq. 22) Eo = + 0.3 V (vs Ag/AgCl) SOD (Cu+) + O2•− +2H+ → SOD (Cu2+) + H2O2 … (Eq. 23) Eo = - 0.2 V (vs Ag/AgCl) Naturally antioxidants, when present, would cause a decrease in O2•− concentration.

1193

Both oxidation and reduction of O2•− could be measured with high sensitivity and selectivity.

1194

Both dismutation products (i.e. H2O2 and O2) could be easily detected simultaneously using

1195

amperometric transducers,115 e.g., the former with an electrodeposited polyprrole covered

1196

glassy carbon microelectrode modified with horseradish peroxidase and the latter with the

1197

same electrode modified with superoxide dismutase enzyme embedded in polymer layers.119

1198

(iii) DNA-based bioelectrosensors worked on the principle of immobilizing (usually calf

1199

thymus−originated) double stranded (ds)-DNA on screen-printed carbon electrodes (SPE), 48 ACS Paragon Plus Environment

Page 49 of 105

Journal of Agricultural and Food Chemistry

1200

followed by the the detection of the guanine oxidation peak between +0.8 and +1.0 V (vs.

1201

Ag/AgCl) by square wave voltammetry. Since the peak current intensity was proportional to

1202

the concentration of DNA base, guanine, the immersion of the DNA-modified electrode into a

1203

Fenton solution (such as Fe(II)+H2O2) produced a signal decrease in the peak current

1204

intensity, whereas the presence of antioxidants restored the signal (demonstrating DNA

1205

integrity) due to hydroxyl radical quenching.115 In order to measure this signal alteration more

1206

effectively, screen-printed carbon was doped with TiO2 nanoparticles, creating a porous

1207

surface structure on which ds-DNA adsorbed better because of specific DNA phosphate-TiO2

1208

interactions.120 A redox mediator, namely tris-2,2’-bipyridine (bipy) ruthenium(II), was

1209

electrooxidized on the electrode surface to subsequently oxidize both the adsorbed ds-DNA

1210

and the antioxidants in solution. Divalent and trivalent ruthenium-bipy species, i.e. RuDNA(II)

1211

and RuDNA(III), represented those redox mediators that were generated in the vicinity of TiO2

1212

nanoclusters. Here, the oxidative damage was produced by [Ru(bipy)3]3+, an efficient oxidant

1213

for the DNA bases, guanine and adenine, that are most sensitive to oxidation. The oxidative

1214

damage on adsorbed DNA was determined by square wave voltammetry via measuring the

1215

current intensity for the oxidation of [Ru(bipy)3]2+ catalyzed by the remaining ds-DNA on the

1216

electrode; antioxidants, when present, had a protective role reducing oxidative damage on

1217

DNA, and were subsequently assayed through competition kinetics by comparing the rate

1218

constants for RuDNA(III) oxidation of antioxidant (in bulk solution) and of adsorbed DNA.120

1219

The recently developed direct current (DC) polarographic assay for AOA estimation is

1220

based on the measurement of anodic current obtained by dropping mercury electrode (DME)

1221

in hydrogen peroxide solution upon the addition of antioxidant compounds. In this DC

1222

polarographic antioxidant assay, the decrease in anodic limiting current of the

1223

[Hg(O2H)(OH)] (hydroxoperhydroxo-mercury(II) complex), formed in alkaline solution of

1224

H2O2, at the potential of mercury oxidation, varied with the concentration of added

49 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 50 of 105

1225

antioxidant capable of H2O2 scavenging, and the method was validated against DPPH and

1226

Folin spectrophotometric assays for wines,121 and for teas and herbal infusions.122 Although

1227

the signal decrease originated from the interaction of antioxidant with hydroperoxo-radicals,

1228

the antioxidant did not directly scavenge H2O2 in the absence of Hg, and the decrease of Hg2+

1229

cathodic current agreed with that of anodic current, leading to the assumption that mercury(II)

1230

reduction caused a decrease in concentration of Hg2+ available for hydroxoperhydroxo-

1231

mercury(II) complex formation, bringing about the decrease in its anodic current.123

1232 1233

2.1.2.3. Nanotechnological Methods

1234 1235

Nanotechnological methods of colorimetric TAC assay usually make use of either the

1236

formation or enlargement of noble metal (Au, Ag, etc.) nanoparticles, abbreviated as AuNPs

1237

or AgNPs, upon reaction of Au(III) or Ag(I) salts with antioxidant compounds. The standard

1238

reduction potentials for Au(III)-Au(0) and Ag(I)-Ag(0) redox couples are 1.5 and 0.8 V,

1239

respectively, and therefore many phenolic antioxidants can be favourably oxidized by

1240

simultaneous reduction of Au(III) and Ag(I) to the corresponding noble metal nanoparticles

1241

(i.e., AuNPs and AgNPs). However, the thermodynamic favourability of these redox reactions

1242

does not guarantee perfect heterogeneous-phase kinetics, and therefore enlargement of

1243

previously formed NP seeds (e.g., citrate-stabilized AgNP seeds) via reaction with

1244

antioxidants is usually preferred, because coating of these NP seeds gives rise to better

1245

linearity of absorbance/concentration responses. The reason that nanomaterial-based methods

1246

have found little use in food science (specifically antioxidant research) is probably the

1247

substoichiometric character of the concerned reduction reactions by antioxidants leading to

1248

NP formation.55 There are also other food constituents (in addition to antioxidants) causing

1249

AuNP or AgNP formation that may give rise to interferences in the assays.

50 ACS Paragon Plus Environment

Page 51 of 105

Journal of Agricultural and Food Chemistry

1250

When AgNPs are dispersed in liquid media, they exhibit a strong UV-Vis absorption

1251

band not present in the spectrum of the bulk metal. This surface plasmon resonance (SPR)

1252

absorption is attributed to the collective oscillation of electrons in the conduction band of

1253

these particles in resonance with the wavelength of incoming light, with a periodic change in

1254

electron density at the surface. The SPR absorption of nano-sized particles having near-zero

1255

dielectric constant gives rise to a localized SPR (LSPR) band. Known LSPR sensors typically

1256

monitor shifts in the peak position or absorption in response to local refractive index changes

1257

in the close vicinity of the NP surface. Although AgNPs have very high molar absorptivities

1258

(ε ≈ 3 × 1011 M−1 cm−1)124 and are expected to allow higher sensitivity in optical detection,

1259

this ε is a corrected value calculated on the basis of the molar amount of nanoparticles per unit

1260

volume of bulk metal. Furthermore, ideal sensitivity in NP-based TAC assays cannot be

1261

achieved because of various factors affecting LSPR absorption such as reaction stoichiometry,

1262

particle size and shape, and dielectric constants of both the metal and the surrounding

1263

medium. Since the properties of surface plasmons can be tailored as a result of altering the NP

1264

surface and, specifically, the shell thickness, the seed-mediated particle growth technique was

1265

adopted (Figures 8 & 9) for developing a AgNPs-based TAC assay.55

1266 1267

Figure 8

1268

Figure 9

1269 1270

Since biothiols play a significant biological role among these compounds due to their strong

1271

reductive ability and capacity to quench reactive oxygen and nitrogen species (ROS/RNS),

1272

and Since the redox potential of the oxidized/reduced forms of glutathione (GSSG/2GSH) is a

1273

basic indicator of the redox environment within a cell, selective quantification of biothiols is

1274

an important topic in bioanalytical chemistry.125 An optical sensor for biologically important

51 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 52 of 105

1275

thiol quantification was designed with the use of Ellman’s reagent (DTNB)-adsorbed gold

1276

nanoparticles (AuNPs) (DTNB-Au-NP) in a colloidal solution.126 5,5’-Dithio-bis(2-

1277

nitrobenzoic acid) (DTNB), a well-documented thiol-selective compound, was adsorbed

1278

through non-covalent interaction onto AuNPs, and the absorbance changes associated with the

1279

formation of the yellow-colored 5-thio-2-nitrobenzoate (TNB2-) anion as a result of reaction

1280

with biothiols was measured at 410 nm (Figure 10). The DTNB reagent could be selectively

1281

desorbed from the derivatized AuNPs surface to give Ellman’s reaction with thiols due to

1282

preferential adsorbabilities of thiols over disulfides127 and to thermodynamic/kinetic

1283

favorability of the thiol-exchange reaction. The linear response of the sensor for cysteine,

1284

glutathione, homocysteine, cysteamine, dihydrolipoic acid and 1,4-dithioerythritol was better

1285

than that of Nile Red dye-derivatized AuNPs sensor for thiol determination.128 Common

1286

biological sample ingredients like amino acids, flavonoids, vitamins, and plasma antioxidants

1287

did not interfere with thiol sensing (Figure 10).126 DTNB-adsorbed AuNPs probes provided

1288

higher sensitivity (i.e., lower detection limits, though not at an order-of-magnitude) in biothiol

1289

determination (Figure 10) than conventional DTNB reagent.110

1290 1291

Figure 10

1292

As H2O2 is a cell membrane-permeable biological oxidant, noble metal nanoparticle-

1293

based methods were also developed to measure H2O2 scavenging antioxidant activity. A gold

1294

nanoshell (GNS)-based optical nanoprobe was developed for assessing hydrogen peroxide

1295

scavenging activity of antioxidants. H2O2 was found to enlarge the AuNPs on the surface of

1296

gold nanoshells (GNS)s precursor nanocomposites (SiO2/AuNPs), and the pre-adsorbed

1297

AuNPs served as nucleation sites for Au deposition. AuNPs on the SiO2 cores were enlarged

1298

by increasing concentrations of H2O2, concomitant with spectral changes corresponding to

1299

AuNP plasmon absorption bands, and antioxidants restricted H2O2-mediated formation of

52 ACS Paragon Plus Environment

Page 53 of 105

Journal of Agricultural and Food Chemistry

1300

GNSs, enabling the determination of their inhibitive activity.129 In another example, H2O2-

1301

induced growth of GNSs was inhibited by the addition of phenolic acids, which affected the

1302

peak wavelength of surface plasmon absorption. Among the tested antioxidants, caffeic acid

1303

was found to be the most efficient H2O2-scavenger, whereas trans-cinnamic acid exhibited the

1304

weakest activity.130

1305 1306

2.1.3. Mixed-Mode (ET‒ and HAT‒Based) Methods

1307 1308

Mixed-mode assays are generally based on the scavenging of a stable radical chromophore

1309

(like ABTS•+ and DPPH•) or fluorophore by antioxidants, in which HAT, ET and PCET

1310

(proton-coupled electron transfer) mechanisms may play different roles to varying extents,

1311

depending on pH, solvent, and other reaction conditions. Schaich and coworkers have

1312

recently directed extensive criticisms to ABTS and DPPH assays131-133 on the grounds that

1313

these assays use sterically hindered, N-centered free radicals as targets to antioxidants rather

1314

than biologically active short-lived radicals (e.g., hydroxyl, superoxide and lipid oxyl radicals

1315

having lifetimes ranging from nano- to ten- seconds) and that their action as radical scavenger

1316

should be irrelevant in vivo.134

1317 1318

2.1.3.1. ABTS/TEAC Assay

1319 1320

ABTS/TEAC assays use intensely-colored cation radicals of ABTS•+ as useful colorimetric

1321

probes accepting hydrogen atoms or electrons supplied by antioxidant compounds. Although

1322

lag-phase assays were preferred at the initial stage of method development, the requirements

1323

for reproducibility and minimizing errors later evolved the assay to a decolorization strategy,

1324

where the initially formed (by H2O2/peroxidase, MnO2 or persulfate) and stabilized ABTS•+

53 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 54 of 105

1325

radical was let to react with the added antioxidant, causing an absorbance decrease at the

1326

characteristic wavelengths. Antioxidant capacity is measured as the ability of the test

1327

compound (e.g., Ph-OH) to decrease ABTS•+ color by intercepting initial oxidation and

1328

preventing ABTS•+ production, or reacting directly with the preformed radical cation. Even

1329

when a fixed-time ABTS assay is preferred, the results may greatly vary for the same

1330

compound (e.g., GSH) depending on the oxidizing agent used to generate the stable colored

1331

radical.33

1332 1333 1334 1335 1336 1337 1338

ABTS + oxidizing agent (such as K2S2O8) → ABTS•+ … (Eq. 24) (λmax=734 nm) ABTS•+ + PhOH → ABTS + PhO• + H+ … (Eq. 25) 2.1.3.2. DPPH• Radical Scavenging Assay

1339 1340

The stable chromogen radical DPPH• was first proposed for quantitating antioxidant content

1341

nearly half a century ago, when Blois used the thiol-containing amino acid cysteine as his

1342

model antioxidant.38 Later it was used as a phenol reagent.135 The more recently introduced

1343

method of Brand-Williams and colleagues136 has been used as a reference point by several

1344

groups of workers.137,138 Reaction with DPPH was adapted for measuring radical quenching

1345

kinetics139,140 and since then numerous variations in methods and time for following the

1346

reaction as well as for calculating relative antioxidant action by reaction stoichiometry have

1347

evolved.131,136 The reaction equation can be formulated with respect to HAT-mechanism,

1348

although proton-coupled ET-mechanism cannot be excluded, especially in phenol-ionizing

1349

solvents:

1350 1351 1352 1353

DPPH• + PhOH → DPPH2 + PhO• … (Eq. 27) where DPPH• is a stable chromogen radical with λmax=515 nm.

1354 54 ACS Paragon Plus Environment

Page 55 of 105

Journal of Agricultural and Food Chemistry

1355 1356

2.1.3.3. DMPD Radical Scavenging Assay

1357 1358

In the presence of ferric iron or reactive species such as hydroxyl radicals, N,N-Dimethyl-p-

1359

phenylenediamine dihydrochloride (DMPD) is converted to the colored DMPD•+ radical

1360

cations, which are scavenged by antioxidant molecules present in test samples, forming the

1361

principle of the DMPD•+ assay. Antioxidant compounds which are able to transfer a hydrogen

1362

atom (or an electron) to DMPD+ cause rapid decolorization of the solution (manifested by an

1363

absorbance descrease at λmax=505 nm) with a stable end-point. The use of DMPD+ has been

1364

widely extended to evaluate the antioxidant capacity of different food products such as fruits,

1365

vegetables and wine.141-143 However, less reproducibility was obtained in the presence of

1366

hydrophobic antioxidants such as tocopherol or BHT.144 Mehdi and Rizvi modified the

1367

DMPD-based method for the measurement of plasma oxidative capacity during human

1368

aging.145 Recently, in a population-based cohort study from Germany, Schöttker et al.

1369

measured the derivatives of reactive oxygen metabolites (d-ROM) in human sera as a proxy

1370

for ROS concentration with the use of DMPD-based d-ROM colorimetric kit.146 Çekiç et al.

1371

simultaneously measured the oxidative status (OS) and antioxidant activity with the aid of a

1372

sensor technique by retaining the pink-colored, positively-charged chromophore of DMPD-

1373

quinone (resulting from the reaction between DMPD and ROS) on a Nafion membrane, where

1374

the 514 nm-absorbance of the sensor membrane was a measure of OS (e.g., derived from

1375

hydroperoxides and labile iron compounds of sera) while antioxidants caused an absorbance

1376

decrease on the membrane due to their ROS scavenging action.147

1377

Other radical probes used for free radical scavenging activity measurement are

1378

Fremy’s salt (galvinoxyl radical: potassium nitrosodisulfonate)148 and the more recently

1379

developed

aroxyl

radical

(2,6-di-tert-butyl-4-(4′-methoxyphenyl)phenoxyl

55 ACS Paragon Plus Environment

radical)

Journal of Agricultural and Food Chemistry

Page 56 of 105

1380

methods,149 but these techniques have been much less frequently preferred than the widely

1381

used ABTS and DPPH assays.

1382 1383

2.1.4. ROS/RNS Scavenging Methods

1384 1385

ROS is a collective term often used to include oxygen radicals [superoxide (O2•-), hydroxyl

1386

(•OH), peroxyl (ROO•) and alkoxyl (RO•)] and certain nonradicals that are either oxidizing

1387

agents and/or are easily converted into radicals, such as HOCl, ozone (O3), peroxynitrite

1388

(ONOO-), singlet oxygen (1O2) and H2O2. RNS is a similar collective term that includes nitric

1389

oxide radical (•NO), ONOO-, nitrogen dioxide radical (•NO2), other oxides of nitrogen and

1390

products arising when NO reacts with O2•-, RO• and ROO•.150 Although ROS and RNS are

1391

essential to human health, an unbalanced excess of these reactive species may cause oxidative

1392

stress‒related diseases.7,151

1393

In ROS/RNS scavenging activity assays, the reactive species generated enzymatically

1394

or by redox-active chemical reagents are allowed to attack a probe, and the subsequent

1395

conversion of this probe is measured spectroscopically or electrochemically, where the extent

1396

of conversion on the probe is a measure of ROS/RNS concentration and its attenuation

1397

indicates the scavenging activity of the tested putative antioxidants. Most conventional

1398

fluorescence (FL) probes for ROS detection react by a free radical mechanism and are rather

1399

non-selectively converted into FL-enhanced or FL-quenched end products, whereas the more

1400

recently developed boronate probes may act more selectively as they undergo nucleophilic

1401

attack by oxidant species to release the hidden fluorescence of a fluorophore containing a

1402

blocking group. Biological macromolecules such as lipids, proteins, sugars and DNA have

1403

been subjected to the attack of biologically relevant reactive species and used as oxidative

1404

stress biomarkers, where various oxidative transformations on these macromolecules have

56 ACS Paragon Plus Environment

Page 57 of 105

Journal of Agricultural and Food Chemistry

1405

been measured. For example, ethane and pentane, conjugated dienes, hydroperoxides,

1406

aldehydes and ketones from lipids; nitro-, chloro-, and bromo-amino acid residues, as well as

1407

carbonyls, -SS-, SOH, and -SOOH compounds, dimers, cross-linked, modified and cleavage

1408

products from proteins; nitrated, chlorinated, brominated and 8-hydroxylated nucleic bases

1409

from DNA can be formed upon oxidation.7,58 In the case of biomarkers, the activity of an

1410

antioxidant can be indirectly mesured by its attenuation of the oxidative hazard formed on the

1411

marker macromolecule. For example, antioxidants may decrease the color intensity of ferric

1412

thiocyanate or ferric-xylenol orange complexes used to measure lipid hydroperoxides, or of

1413

the TBARS chromophore used to measure lipid-derived aldehydes and ketones (e.g.,

1414

malondialdehyde) with questionable specificity.

1415

The selectivity of ROS/RNS assays has been questioned, because frequently there are

1416

more than one reactive species capable of probe conversion, and absorptimetric measurements

1417

in the UV-range (such as the hydrogen peroxide scavenging assay carried out at the intrinsic

1418

maximal absorption wavelength of H2O2 at λ=230 nm)152 suffer from the interference of many

1419

organic compounds absorbing at the same wavelength, thereby yielding inconsistent blanks.

1420

The redundancy of hydroxyl radical scavenging assays have occasionally been discussed in

1421

literature because almost any biological molecule (not necessarily an antioxidant) can react

1422

with this radical at extremely high rates. The possible drawbacks of ROS/RNS scavenging

1423

assays may be listed as follows: (i) if the tested reactive species are produced enzymatically

1424

(e.g., superoxide anion radical by xanthine/xanthine oxidase, reactive species from H2O2 by

1425

peroxidase, HOCl from H2O2 and chloride by myeloperoxidase, reduction of residual nitrate

1426

to nitrite in the course of nitric oxide radical scavenging by NADH-dependent nitrate

1427

reductase), then it is not clear how to differentiate between the ROS/RNS scavenging and

1428

enzyme inhibition actions of antioxidants, and therefore ESR/spin trap methods should

1429

accompany conventional spectroscopic methods in such ambiguous cases; (ii) the tested

57 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 58 of 105

1430

reactive species should not react too rapidly with the selected probe, because in that case, a

1431

wide range of antioxidants reacting with quite different rates cannot be measured. For

1432

example, superoxide radical reacts with the cytochrome c probe much faster than it does with

1433

NBT, rendering the competition of certain antioxidants with the former probe much less

1434

efficient;153 (iii) the tested antioxidant or its oxidation product can enter a direct redox

1435

reaction with the probe via incompletely reacting with the subject ROS/RNS. For example,

1436

ascorbic acid can easily reduce ferricytochrome c in superoxide radical scavenging assay.21

1437

Another example is that in the HOCl scavenging assay in which TNB is oxidized by HOCl to

1438

DTNB, thiol-type antioxidants react with DTNB to produce excessive TNB chromophore;154

1439

(iv) the probe or its conversion product may be unstable, or may itself generate reactive

1440

species during the course of measurement. As a result, such assays should be cautiously

1441

interpreted because they are prone to errors from the above mentioned and other sources,155

1442

and quantification of ROS/RNS scavenging is additionally complicated by the esssentially

1443

non-linear character of either ROS production or consumption with respect to concentration.

1444

Another challenge for the synthesis of future ROS/RNS probe molecules is tailoring them at a

1445

suitable redox potential to specifically quench a given reactive species but not others.

1446 1447

2.1.5. Cellular Antioxidant Activity Assays

1448 1449

Since cellular antioxidant activity assays (CAA) are performed within the cell medium, they

1450

are assumed to better consider certain physico-chemical aspects of the medium such as the

1451

uptake, distribution and metabolism of antioxidants within cells.156 López-Alarcóna and

1452

Denicola157 have recently reviewed cellular antioxidant activity assays in comparison to

1453

classical chemical assays, and to compensate for the possible complexities of antioxidant

1454

action, have recommended the use of CAA to assess the genuine antioxidant activity of a

58 ACS Paragon Plus Environment

Page 59 of 105

Journal of Agricultural and Food Chemistry

1455

compound or extract. To measure CAA, Wolfe and Liu156 used the peroxyl radical oxidation

1456

reaction of the non-fluorescent probe 2’,7’-dichlorofluorescin (DCFH) entrapped in human

1457

hepatocarcinoma HepG2 cells, where the presence of antioxidants attenuated the cellular

1458

fluorescence of the oxidation product, dichlorofluorescein (DCF). This probe may exhibit

1459

several disadvantages such as photochemical instability, incomplete trapping by cells, or

1460

decreased oxidation in the presence of endogenous antioxidants.156 Also, the classical order of

1461

antioxidant effectiveness was not followed in these assays, and the results did not correlate

1462

with those of ORAC.156,158 Halliwell and Whiteman18 directed certain criticisms to cellular

1463

assays, such as interference of enzymic/non-enzymic endogenous antioxidants to the

1464

measurement procedure, intrinsic oxidative stress generation by the cell culture process,

1465

difficulty in distinguishing between intracellular and extracellular fluorescence from chemical

1466

reactions in the culture medium, and inability of DCF fluorescence measurement to

1467

specifically differentiate several reactive species. Effective use of cellular probes also

1468

necessitate a full understanding of the involved mechanistic pathways, environmental factors

1469

(e.g., O2 and pH), distribution and possible intermediary products of the probe, combined with

1470

instrumental artefacts, and actual competition of the measured antioxidants with the probe for

1471

reactive species.159

1472 1473

3. PHYSICO-CHEMICAL ASPECTS OF ANTIOXIDANT ACTION

1474 1475

3.1. Kinetic Solvent Effects and Structure-Activity Relationships

1476 1477

Antioxidant activity/capacity assays should be tested with several reactive species and

1478

oxidizing probes, because the reactivity of known antioxidants toward different ROS/RNS are

1479

quite different. For example, N-acetylcysteine is a powerful scavenger of HOCl, and also

59 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 60 of 105

1480

reacts with hydroxyl radical with a rate constant of 1.36 × 1010 M−1s−1, but only reacts slowly

1481

with H2O2, and does not react at all with superoxide anion radical.160 Antioxidant activity also

1482

strongly depends on the solubility/localization and distribution of antioxidants between

1483

different phases; for example, restricted diffusion of α-tocopherol may reduce its antioxidant

1484

activity in membranes, and a synergistic effect between ascorbic acid and α-tocopherol was

1485

observed under conditions of inhibited peroxidation of linoleate in SDS micelles. A water-

1486

soluble form of α-tocopherol complexed with bovine serum albumin (α-toc:BSA) proved to

1487

be an effective antioxidant for hindering the autoxidation of linoleate in SDS micelles,

1488

whereas (α-toc:BSA) required a long equilibration time with liposomes before α-toc was

1489

transferred to the liposomes to provide effective antioxidant action.161 Antioxidants shown to

1490

be very effective in in vitro TAC assays may exhibit a conflicting order of antioxidant

1491

potency in different systems of varying lipophilic/hydrophilic balance, depending on their

1492

differential abilities to penetrate and interact with the lipid bilayers. A hierarchic order (i.e.

1493

pecking order) of antioxidants in relation to their free radical scavenging ability can be

1494

predicted, based on the comparison of one-electron reduction potentials of the scavenged

1495

reactive species with the corresponding reduction potentials of aryloxy/phenol redox couples.

1496

For example, the formal reduction potentials (at pH=7) of reactive species such as •OH (2.31

1497

V), alkoxyl radical (1.60 V), alkyl peroxyl radical (1.00 V), superoxide anion radical (0.94

1498

V), singlet oxygen (0.65 V), unsaturated fatty acid radical (0.60 V), and H2O2 (0.32 V) are

1499

listed in a comprehensive review.162 Hydrogen peroxide and superoxide anion radical can act

1500

as both an oxidant and reductant, depending on the substrate and medium. Moreover, the

1501

average lifetimes of radicals commonly active in living tissues and in foods vary over ten

1502

orders of magnitude, i.e. •OH (10-9 s), •OR (10-6 s), and ROO• (10 s).132 Thus, disregarding the

1503

distribution between phases, it is natural that different antioxidants would show different

1504

ROS/RNS scavenging abilities based on their redox potentials. In spite of their different

60 ACS Paragon Plus Environment

Page 61 of 105

Journal of Agricultural and Food Chemistry

1505

distribution between lipid membrane and aqueous phases, the fact that vitamin C (ascorbic

1506

acid) can physiologically regenerate vitamin E (α-tocopherol) can be understood by

1507

considering the reduction potentials of α-tocopheroxyl (0.50 V) and ascorbate (0.282 V)

1508

radicals, because α-tocopheroxyl radical can oxidize ascorbate, and is itself converted back to

1509

α-tocopherol. Likewise among flavonoids, quercetin and tea catechins, having reduction

1510

potentials of the corresponding aryloxy radical/phenol couple less than 0.4 V, should be able

1511

to regenerate α-tocopherol from α-tocopheroxyl radical.163 However, it should be borne in

1512

mind that, although a relatively large difference between the standard reduction potentials of

1513

the oxidant and reductant ensures a high equilibrium constant for a nearly complete redox

1514

reaction between the concerned antioxidant and the reactive species it scavenges, it does not

1515

guarantee a high kinetic rate in a given medium, since equilibrium constant is the ratio of the

1516

rate constant of the forward reaction to that of the backward reaction.

1517

The rate and extent of lipid peroxidation can be measured from oxygen uptake,

1518

substrate consumption, or product formation.58 Some secondary oxidation products (e.g.,

1519

hexanal and 2,4-decadienal) or transition metal ions can act as prooxidants and catalyze lipid

1520

oxidation at initial stages, causing early consumption of antioxidants.164 In Cu(II)-induced

1521

lipid peroxidation reactions, the kinetic profile of peroxidation is characterized by three major

1522

parameters: the ‘lag time’ preceding rapid oxidation, the maximal rate of oxidation (Vmax),

1523

and the maximal accumulation of oxidation products (ODmax), and a distinction between

1524

various antioxidants with respect to their mechanism of action can be made based on

1525

observing the different impacts upon these three parameters; for example, antioxidative

1526

effects due to copper-chelating or blockade of copper binding sites can be distinguished from

1527

the effects of free radical quenching.165 The effects of three different flavonoids of similar

1528

structure, i.e. quercetin, morin, and catechin, as potential antioxidant protectors were studied

1529

in a linoleic acid emulsion to yield an inhibitive order of : morin > catechin ≥ quercetin, i.e.

61 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 62 of 105

1530

agreeing with that of formal reduction potentials versus NHE at pH 7, i.e. 0.60, 0.57 and 0.33

1531

V for morin, catechin, and quercetin, respectively. Morin showed antioxidant effect at all

1532

concentrations whereas catechin and quercetin showed both antioxidant and prooxidant

1533

effects depending on their concentrations. The structural requirements for antioxidant activity

1534

in flavonoids interestingly coincided with those for Cu(II)-induced prooxidant activity,

1535

because as the reducing power of a flavonoid increases, Cu(II)–Cu(I) reduction is facilitated

1536

that may end up with the production of reactive species.166

1537

The thermodynamics and kinetics of antioxidant action cannot be properly understood

1538

without understanding solvent effects on HAT- and ET-based reactions, especially on the

1539

latter.167-169 Although HAT-based reactions have been claimed to be relatively rapid at least at

1540

their initial stages, hydrogen bonding in polar solvents may induce dramatic changes in the H-

1541

atom donor activities of phenolic antioxidants and consequently affect the measured reducing

1542

antioxidant capacity.170,171 The rates of oxidation reactions of phenolic compounds (either by

1543

HAT- or proton-coupled electron-transfer (PCET) mechanism) by ROS/RNS are deeply

1544

influenced by H-bond accepting (HBA) and anion solvation abilities of solvents, as well as by

1545

the nature and position of phenol ring substituents (for instance, the rate constants (k, M-1s-1)

1546

for oxidation of phenoxide anions by ClO2 radical, having a standard potential of 0.94 V

1547

versus NHE, were in the descending order of 109, 108, 107, 105 and 103 for resorcinol, p-

1548

methoxyphenol, simple phenol, p-nitrophenol, and p-cyanophenol, respectively, progressively

1549

decreasing with the removal of electron-donating substituents and/or with the increase in

1550

electron-withdrawing substituents on the phenolic ring);172 there is a delicate balance between

1551

the three different, nonexclusive mechanisms of antioxidant action, namely HAT, proton-

1552

coupled ET, and sequential proton loss ET, depending on both the environment and the

1553

reactants.77,173 Choe and Min also discussed the effects of substituents and steric crowding

1554

around the phenolic hydroxyl groups as important parameters determining antioxidant

62 ACS Paragon Plus Environment

Page 63 of 105

Journal of Agricultural and Food Chemistry

1555

activity.174 As an example of the HBA ability of solvent on TAC, in the study of the effects

1556

of polar (e.g., acetonitrile and tert-butyl alcohol) and nonpolar (e.g., cyclohexane) solvents on

1557

the peroxyl-radical-trapping antioxidant activity of some flavonoids, catechol derivatives,

1558

hydroquinone, and monophenols, phenols with two ortho-hydroxyls were the most active

1559

antioxidants, with inhibition rate constants (kinh) in the range of (3−15) × 105 M-1s-1 (in

1560

cyclohexane), whereas in the strong H-bond acceptor solvent tert-butyl alcohol, these rate

1561

constants dramatically declined; however, in the weaker H-bond acceptor solvent acetonitrile,

1562

kinh values were restored close to the values in cyclohexane.175 3,5-Di-tert-butylcatechol, a

1563

very active H-atom donor to DPPH in hexane, showed a dramatic loss of activity in HBA

1564

solvents, especially acetone.176 In general, the aryloxy radicals formed from the oxidation of

1565

catechol (o-dihydroxy phenol) moieties of phenolic compounds are stabilized in non- or weak

1566

hydrogen-bonding solvents by intra-molecular H-bonding, through the interaction of two

1567

adjacent substituents on catechol, i.e., –C(O•)…(HO)C–, bringing about the delocalization of

1568

the odd electron over the whole molecule. An extended delocalization and conjugation of the

1569

π-electrons, enhanced by resonance effects and planarity, favor the lowering of both

1570

ionization potentials (IP) and PhO-H bond dissociation energies (BDE), and affect strongly

1571

the capacity of antioxidants to donate a single electron.177 It should be emphasized that the

1572

relative magnitudes of BDE and IP determine whether the HAT- or ET-mechanism is

1573

predominant for a given PhOH (and these values may show significant variations in polar or

1574

nonpolar solution and gas phases), as a low BDE is required for a strong phenolic antioxidant

1575

essentially acting by H-atom donation whereas a low IP is necessary for a strong antioxidant

1576

functioning essentially via electron-donation.177 The intra-molecular H-bonding stabilization

1577

of the one-electron oxidized catechols will also lower the standard redox potential of the

1578

aryloxy radical/catechol couple, making the phenolic compound a stronger antioxidant in ET-

1579

reactions.173 The strong antioxidant properties of catechol or pyrogallol moieties of

63 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 64 of 105

1580

polyphenols may be predominantly attributed to the intra-molecular H-bonding stabilization

1581

of aryloxy radicals produced from one-electron oxidation of these moieties,178 whereas the

1582

inter-molecular H-bonding abilities of phenolic hydroxyl groups with HBA solvent molecules

1583

lowers antioxidant activity. The antioxidant activities of o-methoxyphenols were decreased in

1584

hydrogen bond accepting media.179 Thus, in evaluating H-atom transfer kinetics of

1585

polyphenols, the ability to form a linear H-bond (with a solvent (S) molecule, in the form of

1586

ArOH…S) should be distinguished from a non-linear H-bond (intra-molecular H-bond),

1587

because only the former may restrict H-atom abstraction from a phenolic compound by a free

1588

radical.180 Most of the PhO-H bond energy differences in H-bonding and non-bonding

1589

solvents (calculated from the enthalpy of the reaction between di-tert-butyl peroxide and

1590

phenol) could be accounted for from the known hydrogen bonding equilibrium between the

1591

solvents and the phenol.181 For strong antioxidant activity, the presence of o-dihydroxy phenol

1592

(catechol) moiety in an antioxidant compound has an additional advantage of chelating

1593

transition metal ions such as iron and copper, thereby hindering transition metal-initiated

1594

Fenton-type reactions generating reactive species. On the other hand, the abnormally high rate

1595

constants in alcohols for H-atom abstraction from 13 hindered and nonhindered phenols by

1596

DPPH• were attributed to the partial ionization of the phenols (Ph-OH) in alcohols and a very

1597

fast electron transfer from phenoxide anion (Ph-O−) to DPPH• ; this also applies for low pKa

1598

phenols in non-hydroxylic polar solvents like di-n-butyl ether, acetonitrile, tetrahydrofurane

1599

and dimethyl sulfoxide.182 The rate constants for DPPH oxidation of phenols in alcohols were

1600

increased by the addition of sodium methoxide and were decreased by added acetic acid. The

1601

initial fast chemistry of the oxidation of curcumin with DPPH• was attributed to the presence

1602

of curcumin anions present at low equilibrium concentrations in alcohols, whereas upon rapid

1603

depletion of these ‘preformed’ anions, ionization of curcumin became partially rate-limiting.77

1604

In this regard, the TEAC coefficients (with respect to the CUPRAC method) of the

64 ACS Paragon Plus Environment

Page 65 of 105

Journal of Agricultural and Food Chemistry

1605

antioxidant compounds: quercetin, catechin, and BHT were higher in pure MeOH than in pure

1606

EtOH, probably due to facilitated electron-transfer in ionizing solvents capable of anion

1607

(phenolate) solvation, because MeOH is the alcohol that best supports ionization.173 The

1608

structure of the B ring in flavonoids seemed to be the primary determinant of antioxidant

1609

activity when studied through fast reaction kinetics with an oxidizing reagent such as

1610

ABTS•+.183 When the H-atom donating ability of 15 flavonoids were studied by ESR, the

1611

second-order reaction rates, primarily governed by O−H bond dissociation energies, were

1612

myricetin > morin > quercetin > fisetin catechin > kaempferol ≈ luteolin > rutin > d-α-

1613

tocopherol > taxifolin > tamarixetin > myricetin 3’,4’,5’-trimethyl ether > datiscetin >

1614

galangin > hesperitin ≈ apigenin.184 Antioxidant effectiveness and reactivity were highly

1615

dependent on the configuration of -OH groups on the flavonoid B and C rings, with very

1616

minor contribution from the A ring. The rate constants for the free radical scavenging action

1617

of flavone, chrysin, and flavonol were low, indicating that the reactivities of 5- and 7-OH

1618

groups at A-ring and 3-OH group at C-ring were very weak and almost negligible; rutin and

1619

quercetin with 3’- and 4’-OH groups at B-ring showed high reactivity, indicating that the o-

1620

dihydroxyl (catechol) structure in the B-ring was the obvious radical target site for

1621

flavonoids.185 Highest reaction rates and stoichiometries were observed with flavonols

1622

capable of being oxidized to ortho-quinones or extended para-quinones.184 The stabilizing

1623

effect of electron-donating groups near phenolic hydroxyls may be exemplified in the much

1624

lower reactivity of 4-methoxy-2,3,5,6-tetramethylphenol (TMMP) than that of α-tocopherol

1625

against peroxyl radicals, because in TMMP, the methoxy group is twisted out of the plane of

1626

the aromatic ring by steric forces and consequently, the p-type lone pair on the methoxyl

1627

oxygen cannot help stabilize the phenoxy1 formed upon abstraction of the phenolic hydrogen,

1628

whereas in tocopherols, the chroman ring system holds the ethereal oxygen’s p-type lone pair

1629

nearly perpendicular to the aromatic ring, thereby providing additional stabilization for the

65 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 66 of 105

1630

resultant phenoxyls.186 The structural requirements for strong antioxidant action with respect

1631

to both thermodynamic efficiency and kinetic rate have been excellently reviewed by Rice-

1632

Evans et al.70 The three criteria for effective radical scavenging of flavonoids were

1633

established as the o-dihydroxy structure in the B ring, the 2,3-double bond in conjugation with

1634

a 4-oxo function in the C ring, and the 3- and 5-OH groups with 4-oxo function in A and C

1635

rings, as characteristically demonstrated in the strong antioxidant flavonoid: quercetin.69,187

1636

Flavanonols and flavanones, due to the lack of conjugation enhancing resonance stabilization

1637

over the entire molecule, are weak antioxidants.163 The half-peak oxidation potentials (Ep/2)

1638

of flavonoids less than 0.2 V are defined as readily oxidizable and therefore good scavengers

1639

of reactive species.163 The reduction potentials (at pH=7) of the aryloxy radical/phenol couple

1640

for quercetin (0.33 V) and myricetin (0.36 V) are lower than those for catechin (0.57 V),

1641

luteolin (0.60 V) and kaempferol (0.75 V), making the former two flavonoids stronger

1642

antioxidants in most in vitro tests. The high antioxidant activities of phenolic and

1643

hydroxycinnamic acids may be attributed to the number and position of phenolic hydroxyls,

1644

the presence of electron-donating substituents (e.g., methoxy groups) in ortho- and para-

1645

position relative to the phenolic –OH thereby stabilizing the produced aryloxy radicals via

1646

oxidation, and the double bonds in side chains. For example, the order of antioxidant

1647

effectiveness of hydroxycinnamates on the induction period of autoxidizing fats was found as:

1648

caffeic > ferulic > p-coumaric acid,188 in accordance with the ET-based CUPRAC results (but

1649

not with the ABTS/TEAC results, in which caffeic acid proved to be less effective than ferulic

1650

and p-coumaric acids).

1651

Recent theoretical analyses of antioxidant action revealed that the kinetics for the free

1652

radical scavenging of polyphenols (formerly assumed to consist of merely HAT and ET

1653

modes) can be further subdivided into three basic mechanisms involving H-atom, proton (H+)

1654

and electron (e−) transfer:189

66 ACS Paragon Plus Environment

Page 67 of 105

Journal of Agricultural and Food Chemistry

1655

(i) Pure HAT and PCET (proton-coupled electron transfer): in pure HAT, the phenolic proton

1656

and electron of the donated H-atom (PhO-H) are transferred to the same atomic orbital of the

1657

free radical whereas PCET involves several molecular orbitals; however, in both HAT and

1658

PCET, the proton and electron are transferred in one kinetic step, shown by the equation.

1659

PhOH + R• → PhO• + RH

1660

In fact, HAT may be visualized as a special case of concerted PCET involving electronically

1661

adiabatic proton transfer. The thermochemistry of PCET and its implications have been

1662

excellently reviewed by Warren et al.190

1663

(ii) ET-PT (electron transfer−proton transfer) is a two-step mechanism started by an e−

1664

transfer and followed by a H+ transfer:

1665

PhOH + R• → PhOH•+ + R−

1666

PhOH•+ + R− → PhO• + RH

1667

In case when PT is very fast, ET-PT is reduced to a HAT process.

1668

(iii) SPL-ET (sequential proton loss−electron transfer), is quite different from HAT, and

1669

occurs (in three consecutive steps) by following the reverse order of ET-PT: it starts with a

1670

proton release (acidic dissociation) forming a phenolate anion, which subsequently transfers

1671

an electron. SPL-ET is favored when the phenolate anion (PhO−) remains stable during the ET

1672

step before reprotonation.

1673

PhOH ↔ PhO− + H+

1674

PhO− + R• → PhO• + R−

1675

R− + H+ → RH

1676

Although these three mechanisms are thermodynamically equivalent (in terms of Gibbs free

1677

energy change), the competition between the different mechanisms is governed by the

1678

kinetics of the limiting step of each mechanism (atom transfer for PCET and electron transfer

1679

for both ET-PT and SPL-ET). By performing quantum chemical calculations on the primary

67 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 68 of 105

1680

roles of the o-dihydroxy catechol moiety and the 3-OH group of quercetin as scavengers of

1681

different types of free radicals, Di Meo et al. reached the conclusion that in nonpolar

1682

environments (e.g., lipid bilayer membranes) and at low pH (e.g., as in the stomach), PCET is

1683

the only possible mechanism, whereas in polar solvents and at pH where quercetin is partly

1684

deprotonated (e.g., in blood plasma), the faster and therefore more predominant process

1685

(SPLET) effectively competes with PCET.189 Thus, the contribution of SPLET to the overall

1686

antioxidant activity of quercetin (also extrapolable to those of other flavonoids) was reported

1687

to increase with a rise in pH where phenols are dissociated to phenolates.189 Amić et al.

1688

theoretically investigated the double-PCET (i.e. 1H+/1e− and 2H+/2e−) processes of free

1689

radical scavenging by flavonoids, and stressed that the significant contribution of the second

1690

PCET mechanism, resulting in the oxidative formation of a quinone/quinone methide, can

1691

effectively distinguish the active flavonoids from inactive ones.191 In other words, the double-

1692

PCET descriptors such as the second O–H bond dissociation enthalpy related to HAT

1693

mechanism in the gas phase and lipid media, and the second electron transfer enthalpy related

1694

to SPLET mechanism in an aqueous (polar) medium were found to better describe the

1695

quantitative structure-activity relationships of the studied set of 21 flavonoids.191 Recently,

1696

Bakhouche et al. theoretically investigated the antioxidant activity of four forms of tocopherol

1697

using the HAT, ET-PT and SPLET mechanisms, and calculated the O-H bond dissociation

1698

free energy (BDFE), ionization potential (IP), proton dissociation free energy (PDFE), proton

1699

affinity (PA) and electron transfer free energy (ETFE) parameters in the gas phase and solvent

1700

media.192 Alpha-tocopherol was shown to be the most reactive form in gas phase with the

1701

lowest BDFE, IP, and PA values. In the gas phase and non-polar media, HAT mechanism was

1702

predominant due to the fact that BDFE was lower than PA and IP, whereas in aqueous and

1703

polar solvents (e.g., DMSO and MeOH), SPLET was more effective because the PA of all

1704

forms of tocopherol was considerably lower than BDFE and IP.192

68 ACS Paragon Plus Environment

Page 69 of 105

Journal of Agricultural and Food Chemistry

1705

In ET-based TAC assays, kinetic solvent effects work in a different way. If the redox

1706

couple of the TAC assay reagent is a coordinatively saturated metal complex (involving

1707

different oxidation states of a given metal ion in the same ligand environment such as

1708

bis(neocuproine)copper(II,I), tris(1,10-phenanthroline)iron(III,II), hexacyanoferrate(III,II))

1709

capable of outer-sphere electron-transfer with the polyphenol,193 then a minor reorientation

1710

(but not substitution) of the already existing ligands around the central metal ion may

1711

expected in the formation of a transient intermediate during the ET process, and consequently,

1712

the reaction rate may only be affected to a limited extent by solvent polarity. In this case, the

1713

magnitude of solvent effects may be determined by the extent of geometric rearrangement

1714

around the coordination center during ET. However, inner-sphere ET reactions of the assay

1715

reagent (e.g., hexaaqua-solvated Fe3+) with phenolics will naturally be affected by the

1716

hydrogen-bonding behavior of the solvent due to stabilization or inhibition of the

1717

intermediary complex formed during electron-transfer. When other factors are disregarded or

1718

assumed to remain constant, HAT-based TAC assays (e.g., ORAC, TRAP, and mixed-mode

1719

ABTS) are generally affected to a greater extent by the solvent behaviour (polarity, HBA,

1720

etc.) than ET-based methods relying on outer-sphere e-transfer (e.g., CUPRAC, ferricyanide,

1721

and FRAP), provided that the ET-reagent chromophore is soluble at effective concentrations

1722

in the solvent of concern.173

1723 1724

3.2. Partitioning of Antioxidants and the Polar Paradox Hypothesis

1725 1726

The capacity for inhibition of lipid peroxidation was measured in many different systems such

1727

as homogeneous solutions, aqueous miscelle dispersions, liposomal membranes, isolated low-

1728

density lipoproteins (LDLs), red blood cells and plasma. Other than the major parameters of

1729

antioxidant activity/capacity, localization of antioxidant, fate of antioxidant-derived radical,

69 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 70 of 105

1730

interaction with other antioxidants, and mobility of antioxidant at the microenvironment are

1731

also important.58

1732

The polar paradox (PP) hypothesis describing the varying behavior of an antioxidant at

1733

different locations can provide limited information about the actually complex behaviour of

1734

antioxidants in oil, lipid and fatty acid emulsions (in water) due to a number of constraints:

1735

(i) The PP hypothesis simply states that due to different locations of polar and nonpolar

1736

antioxidants in bulk oil and water, nonpolar antioxidants should be more effective than their

1737

polar homologues in oil-in-water emulsions because of their better enrichment at the oil-water

1738

interface where oxidation primarily takes place. Even though this was basically confirmed in

1739

earlier cases, recent experiments carried out by varying the polarity of antioxidants by

1740

esterification with various alkyl chain lengths showed that esterified phenolic antioxidants

1741

(such as those of chlorogenic, rosmarinic and dehydrocaffeic acids, tyrosol and

1742

hydroxytyrosol fatty acids, and rutin) essentially obeyed the PP hypothesis up to C8-C12

1743

carbon atoms chain length, beyond which antioxidant activity sharply declined (called the

1744

“cutoff effect”), not conforming to expectations.194-196 Bulky sized phenolic antioxidants

1745

(such as those with long side chains), despite their hydrophobic character, may also show

1746

lower mobility because of steric hindrance, and thus decreased diffusibility toward reactive

1747

centers at the interface.197 It is also probable that the longer chain antioxidants may form

1748

mixed micelles with emulsifiers used to prepare the emulsions resulting in their migration

1749

away from the emulsion droplets.194-196 Emulsifiers may alter the physical location of

1750

antioxidants and the activity of prooxidants; they can also compete with antioxidants for

1751

localization at the interface.197

1752

(ii) The PP hypothesis disregards the possible prooxidant effects of antioxidants under pH-

1753

and concentration-dependent conditions. The concentration dependency is so important that

1754

the hypothesis may only be applicable when the antioxidant reaches a critical concentration so

70 ACS Paragon Plus Environment

Page 71 of 105

Journal of Agricultural and Food Chemistry

1755

that interfacial phenomena are predominant over solubility effects.197 Some polyphenolics

1756

may act as prooxidants within certain concentration ranges by reducing transition metal ions

1757

to their lower oxidation states thereby enhancing Fenton-type oxidation reactions,4 that may

1758

give rise to ‘false antioxidant activity’ measurement with respect to polarity.197

1759

(iii) Excluding surfactant effects, antioxidants can exhibit unexpected interactions with

1760

oxidation promoters and prooxidants (redox reactions) and with trace metal ions

1761

(complexation or chelation), or even among themselves (e.g., through H-bonding and

1762

association), resulting in synergistic or antagonistic effects to change the kinetics of lipid

1763

peroxidation198 and thus deviate from PP hypothetical expectations.

1764

(iv) If the changes in polarity of the phenolic antioxidant is introduced by adding or removing

1765

ring substituents, then such an addition/removal may actually change the O-H bond

1766

dissociation enthalpy (BDE) and hence the antioxidant potency of the molecule.197

1767 1768

3.3. Possible Interactions Among Antioxidant Constituents

1769 1770

The coexistence of multiple antioxidants usually result in additive effects, whereas synergistic

1771

or antagonistic interactions may appear in extreme cases. Synergism or antagonism occur

1772

when the antioxidative protection/scavenging of two antioxidants is greater or smaller than

1773

the sum of their individual effects, respectively. Choe and Min discussed the possible reasons

1774

of synergism in antioxidant activity.174

1775

Synergy and antagonism are two important issues in food and nutrition science

1776

because of the requirement to choose food combinations exhibiting maximal synergism and

1777

minimal antagonism in antioxidant activity. Antioxidative synergy may arise when two

1778

antioxidants (extendable to a greater number of antioxidants) in admixture show the following

1779

behaviors: (i) they display regenerative action, where the primary (stronger) antioxidant is

71 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 72 of 105

1780

regenerated by the weaker (secondary) one; for instance, carotenoids may be regenerated by

1781

α-tocopherol, which in turn may be regenerated by ascorbate, because the order of reduction

1782

potentials for their corresponding oxidized forms (radicals) are: Ecarotenoid ˃ Etocopherol ˃

1783

Eascorbate.162,199 It should be added that this protective action may also suppress the possible

1784

prooxidative action of tocopherol, because the tocopheroxyl radical at relatively high

1785

concentrations with respect to lipids may abstract an H-atom from lipids and produce lipid

1786

radicals, whereas ascorbate may obstruct this pathway by regenerating tocopherol from

1787

tocopheroxyl radical.200 Likewise, inhibited oxygen uptakes of a combination of α-tocopherol

1788

and flavonoids measured in tert-butyl alcohol confirmed the existence of an antioxidant

1789

interaction which produced a more effective inhibition of lipid peroxidation, and the extended

1790

lag-times provided evidence for the regeneration of α-tocopherol by quercetin;201 (ii) they

1791

exhibit cooperative mechanisms of action, for instance, quercetin may act as a metal chelator

1792

while α-tocopherol functions as a radical scavenger.202 On the other hand, although there is no

1793

systematic theory of antagonistic interactions among antioxidant compounds, it is thought to

1794

arise from the regeneration of the minor (less effective) antioxidant from the major (more

1795

effective) one, competition between the formation of antioxidant-radical adducts, and

1796

changing of the microenvironment suitable for one antioxidant in the presence of the other,174

1797

as observed in binary mixtures of α-tocopherol with either caffeic acid or rosmarinic acid, or

1798

in binary mixtures of caffeic acid with either catechin or quercetin.203

1799

From our own laboratory experience with spectrophotometric ET-based TAC assays,

1800

we observed that if antioxidant concentrations in mixtures are appropriately selected to obey

1801

Beer’s law, then the principle of additivity of absorbances (due to the color change of ET-

1802

reagent) would lead to the addivity of total antioxidant capacities of mixture constituents:

1803

TAC(x1, x2 mixture) = TAC(x1) + TAC(x2) in mmol (or micromol) trolox-equiv./L

72 ACS Paragon Plus Environment

Page 73 of 105

Journal of Agricultural and Food Chemistry

1804

Only in this case, real synergistic or antagonistic interactions (if any) would manifest

1805

themselves. If the results of spectrophotometric ET-based (e.g., CUPRAC, FRAP,

1806

ferricyanide) or mixed-mode (e.g., ABTS and DPPH radical scavenging) assays relying on a

1807

fixed-time end-point are evaluated with the above approach, one would see that most data

1808

assumed to originate from synergistic or antagonistic interactions (via calculations involving

1809

quasi-linear combinations of lag-time or inhibition percentage) are in fact additive,11,64

1810

because in accordance with Beer’s law, absorbance varies linearly with concentration within a

1811

reasonable range, which is not valid for the essentially nonlinear parameters of lag-time or

1812

inhibition percentage;204,205 The synergy observed with the CUPRAC method in (BHT+BHA)

1813

or (BHT+TBHQ) mixtures173 has not yet been fully interpreted, but thought to arise from

1814

enhanced solubilization and stabilization of the semi-quinone radical adducts formed in the

1815

course of oxidation via inter-molecular H-bonding and dimerization. Thiol mixtures

1816

seemingly exhibiting synergy in FRAP and ABTS assays may involve further oxidations in

1817

the presence of multiple thiols (e.g., the partially reversible oxidation reaction expected to

1818

generate disulfides may go further beyond that stage, giving rise to sulfenic and sulfinic

1819

acids).

1820

Synergy and antagonism still remain to be hardly quantifiable behaviors of diverse

1821

antioxidant mixtures. Recently, extensive efforts are being spent for quantifying synergy or

1822

antagonism in antioxidant activity using different reagents and calculation methods. For

1823

instance, Skroza et al. observed synergy in the binary mixtures of resveratrol with catechin

1824

and gallic acid.206 Prieto et al. used a dose-dependent mathematical model based on

1825

probability functions, where the interactive effects between antioxidants were introduced into

1826

the model with simple auxiliary functions that describe the parametric variations induced by

1827

each antioxidant in the presence of the other and a collective index of parametric responses

73 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 74 of 105

1828

was generated, and experimentally demonstrated this effect using DPPH and ABTS assays,

1829

extendable to other ET-assays.207

1830 1831

3.4. Enhanced Solubilization of Antioxidants via Coupled Oxidation Using TAC

1832

Reagents

1833 1834

Since dietary fibre-phenolic compounds (DF-PC) are made up of bound hydroxycinnamic

1835

acids, TAC of cereal fibre has been disputed, and therefore the actual TAC of DF-PC has

1836

been largely underestimated because of their low water and organic solvent solubility. In fact,

1837

a proper assessment of DF-PC antioxidant capacity requires a multiple-step extraction and an

1838

appropriate chemical hydrolysis to release PC and to permit them to exert antioxidant activity

1839

in the in vitro assays. For many years, it was not clear whether the water insoluble DF-PC

1840

fraction could exert any antioxidant action itself, i.e. without any chemical hydrolysis.208

1841

Serpen et al.209 were the first researchers to demonstrate the concept of antioxidant activity of

1842

insoluble material measured in a different way. In reality, the slow and continuous release of

1843

DF-PC from the insoluble material (known to survive for a considerable time) in the human

1844

gastrointestinal tract has been established to occur, and in particular, DF-PC may favorably

1845

act in vivo quenching the soluble radicals that are continuously formed in the intestinal

1846

tract.208 Tufan et al.210 determined the TAC of cereals (i.e. barley, wheat, rye, oat) by the

1847

‘QUENCHER procedure’ (involving forced solubilization of bound phenolics by oxidizing

1848

TAC reagents) with the direct use of copper(II)-neocuproine (Cu(II)-Nc) reagent of the

1849

CUPRAC assay; the assay operated without a need for completely solubilizing or extracting

1850

antioxidant constituents of insoluble matrices before the CUPRAC reaction, because the

1851

driving force for the CUPRAC oxidation of bound phenolics was greater than that for their

1852

solubilization, making the whole coupled-oxidation process thermodynamically favourable.

74 ACS Paragon Plus Environment

Page 75 of 105

Journal of Agricultural and Food Chemistry

1853

The authors effectively measured the TAC values of cereals by their proposed QUENCHER-

1854

CUPRAC assay, and their results linearly correlated with those of reference methods (ABTS

1855

and DPPH). They found additive responses of polyphenolic mixtures in a cellulose matrix

1856

with their proposed method.210 Solvent effects in the QUENCHER approach were recently

1857

discussed.211 Enhanced solubilization of antioxidant compounds via coupled oxidation is an

1858

active area of research, with many potential contributions in food analytical chemistry that

1859

may allow the building of unique databases for determining the antioxidant capacity of foods

1860

having insoluble moieties, e.g., the polysaccharide matrix.212

1861 1862

4. A PRACTICAL GUIDE TO ANTIOXIDANT ASSAY SELECTION

1863 1864

In conclusion, we also propose a rather subjective guide (primarily stemming from our

1865

laboratory’s experiences) for assay selection, briefly summarizing which assay can be chosen

1866

(or not to be preferred) to serve a specific purpose. Ideally, the chosen test should be

1867

sensitive, selective, robust, reproducible, use conventionally available reagents and

1868

instruments, and measure a wide variety of antioxidant types including both lipophilic and

1869

hydrophilic antioxidants, but unfortunately these criteria are not met by a single specific

1870

assay. It is obvious that one has to decide at the start whether antioxidant activity (basically

1871

reaction rate) or antioxidant capacity (stoichiometric conversion efficiency) is to be measured.

1872

Antioxidant activity tests having an area-under-curve (AUC) approach toward fluorescence

1873

decay of a probe attacked by reactive species in the presence of antioxidants (like ORAC) can

1874

simultaneously measure the lag phase, initial rate, and total extent of inhibition of

1875

antioxidants. On the other hand, such tests may not differentiate between reaction rate and

1876

radical scavenging efficiency, and may give positively-biased results for slowly reacting

1877

antioxidants. It is also recommended that test probe selection should be made in accordance

75 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 76 of 105

1878

with the classical definition of an ‘antioxidant’, i.e. the concentration of the tested antioxidant

1879

should be much smaller than that of the probe, a requirement which is not strictly obeyed in

1880

certain HAT-based assays using fluorescent probes.

1881

(a) The working pH is an important parameter of antioxidant assay selection. FRAP,

1882

CUPRAC and Folin methods can be used for acidic (pH 3.6), neutral (pH 7.0) and alkaline

1883

(pH 10) media measurements, respectively. It should be borne in mind that phenolic

1884

antioxidants are undissociated, partly dissociated, and predominantly dissociated at the

1885

working pH values of FRAP, CUPRAC and Folin methods, respectively, possibly reflecting

1886

an underestimation (in the case of FRAP) or overestimation (in the case of Folin) of the in

1887

vitro antioxidant action simulating physiological conditions. Classical ‘ferric reducing assay’

1888

utilizing ferricyanide incubation with the antioxidant followed by ferric ion addition at the

1889

end of the reaction (prior to color development) works ar pH 6.6, while modified ferricyanide

1890

assay initially incorporating Fe(III) in the incubation solution has a pH of 1.7. A mixed-mode

1891

(HAT/ET) free radical scavenging assay like DPPH may also be very sensitive to pH, as the

1892

rate-determining step of proton-coupled electron transfer may be the acidic dissociation of a

1893

phenolic proton, followed by a fast electron-transfer from the phenolate anion to DPPH. In

1894

general, a rise in pH significantly enhances both the rate and efficiency of phenolics oxidation

1895

in ET- and mixed-mode assays, owing to deprotonation.

1896

(b) Use of natural materials in the selected assay may adversely affect the reproducibility of

1897

results. For example, the natural pigment, crocin, or the natural protein, β-phycoerythrin, may

1898

contain impurities and may vary from lot-to-lot in production. The same applies for lipids

1899

(e.g., purified LDL) in testing antioxidative action against lipid peroxidation.

1900

(c) Multi-charged chromophores of TAC redox probes strongly interact with solvent water

1901

molecules by ion-dipole interaction, and therefore respond less to hydrophobic antioxidants.

1902

CUPRAC and ABTS methods, having mono-positive charged chromophores, can

76 ACS Paragon Plus Environment

Page 77 of 105

Journal of Agricultural and Food Chemistry

1903

simultaneously measure hydrophilic and lipophilic antioxidants, while ORAC, FRAP,

1904

ferricyanide and Folin methods having either hydrophilic or multi-charged chromophores

1905

require the use of cyclodextrins (CDs) or micellar solutions to enhance the solubilization of

1906

lipophilics. Unfortunately, cyclodextrins form inclusion complexes with antioxidants, which

1907

may adversely affect subsequent reaction with a redox probe if antioxidant-CD complex is too

1908

stable at relatively high concentrations of CDs necessarily used for solubilization.

1909

(d) Colorimetric reagents having chromophores absorbing distinctly in the visible range of the

1910

electromagnetic spectrum (i.e. as far as possible to the UV range) are the appropriate methods

1911

of choice to deal with the background color of plant pigments and other UV-absorbing

1912

substances. The background color arising from the matrix shows maximal adverse effects on

1913

the precision of color-fading reactions (like DPPH and ABTS) rather than on that of color-

1914

forming reactions (like FRAP, CUPRAC, ferricyanide). Antioxidant activity assays based on

1915

diene formation or hydrogen peroxide scavenging, all involving UV measurements, may be

1916

adversely affected from UV-absorbing interferents. This applies well to most plant extracts

1917

and pharmaceutical preparates, where many constituents strongly absorb in the UV range.

1918

(e) Redox potential of the selected probe is important in antioxidant assay selectivity.

1919

CUPRAC, ABTS and FRAP methods use oxidant probes having 0.60, 0.68 and 0.70 V

1920

reduction potentials (vs NHE), which are at the right potential to oxidize many common

1921

antioxidants lying in the 0.2-0.6 V redox potential range, while conventional ferricyanide test

1922

has an insufficient potential (0.36 V vs NHE) to oxidize certain antioxidants and Folin test has

1923

an indefinitely high potential (enabling the oxidation of citric acid, reducing sugars and some

1924

amino acids, which are not ‘true’ antioxidants). The newly developed colorimetric redox

1925

reagents containing Ce(IV), Cr(VI) and Mn(VII) centers have to be tested for longer periods

1926

of time to see the exact interference results, but it may be speculated that their high redox

77 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 78 of 105

1927

potentials, even when restricted by certain measures, would not enable highly selective TAC

1928

assays as other organics would be co-oxidized.

1929

(f) When metal ion‒containing probes (such as those of FRAP and CUPRAC) are used,

1930

strong chelating or complexing agents may interfere with the assay. For example, we have

1931

observed that EDTA at sufficiently low concentrations to preserve serum samples does not

1932

interfere with the CUPRAC assay, but at higher concentrations, it may partly displace the

1933

neocuproine ligand from the coordination sphere of Cu(II) and decrease the redox potential of

1934

the Cu(II,I) couple, thereby preventing the oxidation of certain antioxidants. The same applies

1935

for FRAP when strongly complexing ligands displace the tripyridyltriazine (TPTZ) ligand

1936

from the coordination sphere of Fe(III) ion.

1937

(g) Metal ion‒containing probes may also suffer from undesired redox cycling of

1938

antioxidants, causing serious errors in TAC calculation. For example, the relative

1939

concentration of free Fe3+ ion in the FRAP reagent is higher than stoichiometrically required

1940

for the 1:2 Fe(III)-TPTZ complex. As the close standard potentials of Fe3+,2+ and

1941

FeIII,II(TPTZ)2 redox couples are 0.77 V and 0.70 V, respectively, significant amunts of Fe2+

1942

may be formed along with FeII(TPTZ)2 upon reduction reaction with antioxidants, and may

1943

subsequently give rise to Fenton-type reactions with H2O2 or dissolved O2. Production of

1944

reactive species may give rise to redox cycling of antioxidants causing erroneous TAC results.

1945

On the other hand, the standard potentials of Cu2+,1+ and CuII,I(Nc)2 redox couples are quite

1946

different in magnitude, being 0.17 V and 0.60 V, respectively, and therefore no redox cycling

1947

is observed because the reduction product of the CUPRAC reagent with antioxidants is

1948

definitely CuI(Nc)2, which may not be reoxidized with H2O2.

1949

(h) The active constituent of FRAP reagent is the ferric-TPTZ complex bearing high-spin iron

1950

which exhibits slow kinetics, and therefore FRAP does not sufficiently respond to thiols

1951

mainly due to kinetic rather than thermodynamic reasons. As a result, due to its inefficiency

78 ACS Paragon Plus Environment

Page 79 of 105

Journal of Agricultural and Food Chemistry

1952

in oxidizing biothiols (including both small-molecule and protein thiols), FRAP method may

1953

not be recommended for biological samples. We have observed in our laboratory that

1954

CUPRAC and ABTS methods give satisfactory results for measuring the TAC values of

1955

serum samples, due to their capability of simultaneously measuring lipophilic and hydrophilic

1956

serum antioxidants together with thiols. The results of ABTS test for biothiols should be

1957

carefully interpreted, because the TEAC coefficients of ABTS test for biothiols vary roughly

1958

between 1.2-1.6 depending on the method of ABTS•+ radical preparation, hinting to the fact

1959

that thiol (RSH) oxidation in ABTS assay proceeds further beyond disulfides (i.e. RSSR,

1960

encountered in CUPRAC test with a TEAC coefficient of roughly 0.5 for RSH) to higher

1961

oxidation products.

1962

(i) Structural limitations may be responsible for the slow kinetics of phenolic antioxidants

1963

having bulky substituents. For example, mixed-mode assays using ABTS and DPPH hindered

1964

radicals may suffer from steric accessibility problems caused by polymeric phenols having

1965

multiple hydroxyl groups and ring adducts (e.g., tannins). Use of assays using outer-sphere

1966

electron-transfer agents (such as CUPRAC, FRAP and ferricyanide) may be more appropriate

1967

in such samples. It should always be remembered that reaction kinetics is a function of

1968

antioxidant structure, pH, temperature and solvent type, and many ET- (e.g., FRAP) or

1969

mixed-mode (ABTS and DPPH) assays cannot go to completion within the protocol time of

1970

the assay.

1971

(j) In competitive assays, one has to avoid ‘short-circuit’ reactions. The tested antioxidant

1972

may react directly with the probe (e.g., thiol-NBT reaction in superoxide scavenging test)

1973

instead of competing with the radical. Similar reactions may occur in enzyme-mediated

1974

generation of reactive species (e.g., peroxidase may directly oxidize ABTS in hydrogen

1975

peroxide scavenging test without adding H2O2).

79 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 80 of 105

1976

(k) Several assays, preferably working in different modes (i.e. HAT, ET, mixed-mode), may

1977

be used together to reveal the whole picture of antioxidative action. It should be borne in

1978

mind that a fast-reacting antioxidant may stay behind a slow-reacting one in antioxidant

1979

ranking when an end-point assay is used because of the fact that the latter quenches a greater

1980

number of free radicals per molecule. Thus it may be advisable to use both AOA and TAC

1981

assays in biologically relevant antioxidant evaluation.

1982 1983

ACKNOWLEDGMENTS

1984 1985

The authors would like to express their gratitude to Istanbul University-Application &

1986

Research Center for the Measurement of Food Antioxidants (Istanbul Universitesi Gida

1987

Antioksidanlari Olcumu Uygulama ve Arastirma Merkezi). One of the authors (R. Apak)

1988

wishes to thank the Istanbul University Research Fund (BAP) for sponsoring his participation

1989

in the Pittcon Analytical Chemistry Meeting (8-12 March, 2015; New Orleans, USA) under

1990

the research project UDP-51475.

1991 1992 1993 1994 1995 1996 1997 1998 1999 2000 2001 2002 2003 2004 2005 2006 2007 2008 2009

LIST OF ABBREVIATIONS AAPH = 2,2’azobis (2-methylpropionamidine) dihydrochloride ABAP = 2,2'-azobis-(2-amidinopropane) dihydrochloride ABTS = 2,2’-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid AOA = antioxidant activity AOX = antioxidants Ag-NPs = silver nanoparticles AUC = area under curve Au-NPs = gold nanoparticles β-PE = β-phycoerythrin CERAC = Ce(IV) reducing antioxidant capacity CHROMAC = Cr(VI) reducing antioxidant capacity CL = Chemiluminescence CUPRAC = cupric reducing antioxidant capacity Cu(II)-Nc = bis (neocuproine) copper(II) cation [Cu(Nc)2]+ = cuprous neocuproine 80 ACS Paragon Plus Environment

Page 81 of 105

2010 2011 2012 2013 2014 2015 2016 2017 2018 2019 2020 2021 2022 2023 2024 2025 2026 2027 2028 2029 2030 2031 2032 2033 2034 2035 2036 2037 2038 2039 2040 2041 2042 2043 2044 2045 2046 2047 2048 2049 2050 2051 2052 2053 2054 2055 2056 2057 2058 2059

Journal of Agricultural and Food Chemistry

CV = cyclic voltammetry DCM = dichloromethane DHBA = dihydroxybenzoic acid DMPD = N,N-dimethyl-p-phenylenediamine dihydrochloride DPPH = 2,2-di(4-tert-octylphenyl)-1-picrylhydrazyl DPV = differential pulse voltammetry DTNB = 5,5’-Dithio-bis(2-nitrobenzoic acid) TNB = 5-Thio-2-nitrobenzoate ECL = electrochemiluminescence EPR = electron paramagnetic resonance ESR = electron spin resonance ET = electron transfer FRAP = ferric reducing antioxidant power GAE = gallic acid-equivalent GC = glassy carbon GSH-Px = glutathione peroxidase GSH-Rx = glutathione reductase HAT = hydrogen atom transfer H2O2 = hydrogen peroxide HOCl = hypochlorous acid HPS = hydrogen peroxide scavenging LMWA = low molecular-weight antioxidants LSPR = localized surface plasmon resonance KMBA = -keto-γ-methiolbutyric acid M-β-CD = methyl-β-cyclodextrin NBT = nitroblue tetrazolium Nc = neocuproine NO = nitric oxide radical NO = nitrogen dioxide radical 2 1 O2 = singlet oxygen O2− = superoxide anion radical OH = hydroxyl radical ONOO− = peroxynitrite anion ORAC = oxygen radical absorbance capacity PCA = principal component analysis PLS = partial least squares PP = polar paradox RO = alkoxyl radical ROO = peroxyl radical ROS = reactive oxygen species RNS = reactive nitrogen species SOD = superoxide dismutase SPE = screen-printed carbon electrode SPR = surface plasmon resonance SRSA = superoxide radical scavenging activity SWV = square-wave voltammetry TAC = total antioxidant capacity TBARS = thiobarbituric acid-reactive substances TBHQ = tert-butylhydroquinone TE = trolox-equivalent 81 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

2060 2061 2062 2063 2064 2065 2066 2067 2068 2069 2070 2071 2072 2073 2074 2075 2076 2077 2078 2079 2080 2081 2082 2083 2084 2085 2086 2087 2088 2089 2090 2091 2092 2093 2094 2095 2096 2097 2098 2099 2100 2101 2102 2103 2104 2105 2106 2107

Page 82 of 105

TEAC = trolox-equivalent antioxidant capacity TOSC = total oxyradical scavenging capacity TPTZ = 2,4,6-tripyridyl-s-triazine TRAP = total peroxyl radical trapping antioxidant parameter XOD = xanthine oxidase

REFERENCES (1) Trevisan, M.; Browne, R.; Ram, M.; Freudenheim, J.; Carosella, A. M.; Armstrong, D. Correlates of markers of oxidative status in the general population. Am. J. Epidemiol. 2001, 154, 348-356. (2) Prior, R. L.; Wu, X.; Schaich, K. Standardized methods for the determination of antioxidant capacity and phenolics in foods and dietary supplements. J. Agric. Food Chem. 2005, 53, 4290–4302. (3) Taverniers, I.; De Loose, M; Van Bockstaele, E. Trends in quality in the analytical laboratory. II. Analytical method validation and quality assurance. Trends Anal. Chem. 2004, 23, 535-552. (4) Niki, E.; Noguchi, N. Evaluation of antioxidant capacity. What capacity is being measured by which method? IUBMB Life 2000, 50, 323-329. (5) Frankel, E. N.; Meyer, A. S. The problems of using one dimensional methods to evaluate multifunctional food and biological antioxidants. J. Sci. Food Agric. 2000, 80, 1925-1941. (6) Halliwell, B. Antioxidant characterization. Methodology and mechanism. Biochem. Pharmacol. 1995, 49, 1341-1348. (7) Niki, E. Assesment of Antioxidant Capacity in vitro and in vivo. Free Radical Biol. Med. 2010, 49, 503-515. (8) Özyürek, M.; Güçlü, K.; Apak, R. The main and modified CUPRAC methods of antioxidant measurement. Trends Anal. Chem. 2011, 30, 652-664. (9) Aruoma, O. I. Methodological considerations for characterizing potential antioxidant actions of bioactive components in plant foods. Mutat. Res. 2003, 523-524, 9-20. (10) Wayner, D. D. M.; Burton, G. W.; Ingold, K. U.; Locke, S. Quantitative measurement of the total peroxyl radical trapping antioxidant capability of human blood plasma by controlled peroxidation. The important contribution made by plasma proteins. FEBS Lett. 1985, 187, 33-37. (11) Demirci Çekiç, S.; Kara, N.; Tütem, E.; Sözgen Başkan, K.; Apak, R. Protein−incorporated serum total antioxidant capacity measurement by a modified CUPRAC (CUPric Reducing Antioxidant Capacity) method. Anal. Lett. 2012, 45, 754-763. (12) Özyürek, M.; Bektaşoğlu, B.; Güçlü, K.; Apak, R. Hydroxyl radical scavenging assay of phenolics and flavonoids with a modified cupric reducing antioxidant capacity (CUPRAC) method using catalase for hydrogen peroxide degradation. Anal. Chim. Acta 2008, 616, 196-206. (13) Demirci Çekiç, S.; Çetinkaya, A.; Avan, A. N.; Apak, R. Correlation of total antioxidant capacity with reactive oxygen species (ROS) consumption measured by oxidative conversion, J. Agric. Food Chem., 2013, 61, 5260-5270. (14) Huang, D.; Ou, B.; Prior, R. L. The chemistry behind antioxidant capacity assays J. Agric. Food Chem. 2005, 53, 1841-1856.

82 ACS Paragon Plus Environment

Page 83 of 105

2108 2109 2110 2111 2112 2113 2114 2115 2116 2117 2118 2119 2120 2121 2122 2123 2124 2125 2126 2127 2128 2129 2130 2131 2132 2133 2134 2135 2136 2137 2138 2139 2140 2141 2142 2143 2144 2145 2146 2147 2148 2149 2150 2151 2152 2153 2154 2155 2156 2157

Journal of Agricultural and Food Chemistry

(15) Magalhaes, L. M.; Segundo, M. A.; Reis, S.; Lima, J. L. F. C. Methodological aspects about in vitro evaluation of antioxidant properties. Anal. Chim. Acta 2008, 613, 1-19. (16) Halliwell, B. How to characterize a biological antioxidant. Free Radical Res. Commun. 1990, 9, l-32. (17) Gutteridge, J. M. C. Lipid peroxidation and antioxidants as biomarkers of tissue Damage. Clin. Chem. 1995, 41, 1819-1828. (18) Halliwell, B.; Whiteman, M. Measuring reactive species and oxidative damage in vivo and in cell culture: how should you do it and what do the results mean? Br. J. Pharmacol. 2004, 142, 231-255. (19) Finley, J. W.; Kong, A. N.; Hintze, K. J.; Jeffery, E. H.; Ji, L. L.; Lei, X. G. Antioxidants in foods: State of the science important to the food industry. J. Agric. Food Chem. 2011, 59, 6837-6846. (20) Madhavi, D. L.; Deshpande, S. S.; Salunkhe, D. K. Introduction. In Food Antioxidants: Technological, Toxicological, and Health Perspectives; Madhavi, D. L., Deshpande, S. S., Salunkhe, D. K., Eds.; Marcel Dekker, Inc.: New York, 1996; pp. 1-4. (21) Halliwell, B.; Murcia, M. A.; Chirico, S.; Aruoma, O. I. Free radicals and antioxidants in food and in vivo: what they do and how they work. Crit. Rev. Food Sci. Nutr. 1995, 35, 7-20. (22) Bartosz, G. Non-enzymatic antioxidant capacity assays: Limitations of use in biomedicine. Free Radical Res. 2010, 44, 711-720. (23) Shahidi, F.; Ambigaipalan, P. Phenolics and polyphenolics in foods, beverages and spices: Antioxidant activity and health effects – A review, J. Funct. Foods 2015, 18, 820-897. (24) Shahidi, F.; Zhong, Y. Measurement of antioxidant activity, J. Funct. Foods 2015, 18, 757-781. (25) Amorati, R.; Valgimigli, L. Advantages and limitations of common testing methods for antioxidants, Free Radical Res. 2015, 49, 633-649. (26) Wayner, D. D. M.; Burton, G. W.; Ingold, K. U.; Barclay, L. R.; Locke, S. J. The relative contributions of vitamin E, urate, ascorbate and proteins to the total peroxyl radical-trapping antioxidant activity of human blood plasma. Biochim. Biophys. Acta. 1987, 924, 408-419. (27) Bors, W.; Heller, W.; Michel, C.; Saran, M. Flavonoids as antioxidants: Determination of radical scavenging efficiencies. Methods Enzymol. 1990, 186, 343355. (28) Tubaro, E.; Ghiselli, A.; Rapuzzi, E.; Maiorino, M.; Ursini, E. Analysis of plasma antioxidant capacity by competition kinetics. Free Radical Biol. Med. 1998, 24, 1228-1234. (29) Cao, G.; Alessioo, H. M.; Cutler, R. G. Oxygen-radical absorbance capacity assay for antioxidants. Free Radical Biol. Med. 1993, 14, 303-311. (30) Ou, B.; Hampsch-Woodill, M.; Prior, R. L. Development and validation of an improved oxygen radical absorbance capacity assay using fluorescein as the fluorescent probe. J. Agric. Food Chem. 2001, 49, 4619-4626. (31) Winston, G. W.; Regoli, F.; Dugas, A. J. J.; Fong, J. H.; Blanchard, K. A. A rapid gas chromatographic assay for determining oxyradical scavenging capacity of antioxidants and biological fluids. Free Radical Biol. Med. 1998, 24, 480-493. (32) Regoli, E. Total oxyradical scavenging capacity (TOSC) in polluted and translocated mussels: A predictive biomarker of oxidative stress. Aquat. Toxicol. 2000, 50, 351-361. 83 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

2158 2159 2160 2161 2162 2163 2164 2165 2166 2167 2168 2169 2170 2171 2172 2173 2174 2175 2176 2177 2178 2179 2180 2181 2182 2183 2184 2185 2186 2187 2188 2189 2190 2191 2192 2193 2194 2195 2196 2197 2198 2199 2200 2201 2202 2203 2204 2205

Page 84 of 105

(33) Bartosz, G. Total antioxidant capacity. In Advances in Clinical Chemistry, 1st Edition; Spiegel, H. E., Ed.; Academic Press: New York, 2003; Vol. 37, pp. 219292. (34) Folin, O.; Ciocalteu, V. On tyrosine and tryptophane determinations in proteins. J. Biol. Chem. 1927, 73, 627-650. (35) Singleton, V. L.; Orthofer, R.; Lamuela-Raventos, R. M. Analysis of total phenols and other oxidation substrates and antioxidants by means of folin-ciocalteu reagent. Methods Enzymol. 1999, 299, 152-178. (36) Pellegrini, N.; Re, R.; Yang, M.; Rice-Evans, C. Screening of dietary carotenoids and carotenoid-rich fruit extracts for antioxidant activities applying 2,2'azinobis (3- ethylenebenzothiazoline-6-sulfonic acid) radical cation decolorization assay. Methods Enzymol. 1999, 299, 379-389. (37) Re, R.; Pellegrini, N.; Proteggente, A.; Pannala, A.; Yang, M.; Rice-Evans, C. Antioxidant activity applying an improved ABTS radical cation decolorization assay. Free Radical Biol. Med. 1999, 26, 1231-1237. (38) Blois, M. S. Antioxidant determinations by the use of a stable free radical. Nature 1958, 181, 1199-1200. (39) Sanchez-Moreno, C.; Larrauri, J. A.; Saura-Calixto, F. A. A procedure to measure the antiradical efficiency of polyphenols. J. Sci. Food Agric. 1998, 76, 270276. (40) Apak, R.; Güçlü, K.; Özyürek, M.; Karademir, S. E. A novel total antioxidant capacity index for dietary polyphenols, vitamins C and E, using their cupric ion reducing capability in the presence of neocuproine: CUPRAC method. J. Agric. Food Chem. 2004, 52, 7970-7981. (41) Apak, R.; Güçlü, K.; Özyürek, M.; Karademir, S. E.; Altun, M. Total antioxidant capacity assay of human serum using copper(II)-neocuproine as chromogenic oxidant: the CUPRAC method. Free Radical Res. 2005, 39, 949-961. (42) Benzie, I. F. F.; Strain, J. J. The ferric reducing ability of plasma (FRAP) as a measure of ‘antioxidant power’: The FRAP assay. Anal. Biochem. 1996, 239, 70-76. (43) Benzie, I. F.; Strain, J. J. Ferric reducing/antioxidant power assay: Direct measure of total antioxidant activity of biological fluids and modified version for simultaneous measurement of total antioxidant power and ascorbic acid concentration. Methods Enzymol. 1999, 299, 15-27. (44) Benzie, I. F. F.; Szeto, Y. T. Total antioxidant capacity of teas by the ferric reducing/antioxidant power assay. J. Agric. Food Chem. 1999, 47, 633-636. (45) Pulido, R.; Bravo, L.; Saura-Calixto F. Antioxidant activity of dietary polyphenols as determined by a modified ferric reducing/antioxidant power assay. J. Agric. Food. Chem. 2000, 48, 3396-3402. (46) Oyaizu, M. Studies on products of browning reaction: antioxidative activity of products of browning reaction prepared from glucosamine. Jpn. J. Nutr. 1986, 44, 307-315. (47) Berker, K. I.; Güçlü, K.; Tor, I.; Demirata Öztürk, B.; Apak, R. Total antioxidant capacity assay using optimized ferricyanide / Prussian blue method. Food Anal. Methods 2010, 3, 154-168. (48) Berker, K. I.; Güçlü, K.; Tor, I.; Apak, R. Comparative evaluation of Fe(III) reducing power-based antioxidant capacity assays in the presence of phenanthroline, batho-phenanthroline, tripyridyltriazine (FRAP), and ferricyanide reagents. Talanta 2007, 72, 1157-1165.

84 ACS Paragon Plus Environment

Page 85 of 105

2206 2207 2208 2209 2210 2211 2212 2213 2214 2215 2216 2217 2218 2219 2220 2221 2222 2223 2224 2225 2226 2227 2228 2229 2230 2231 2232 2233 2234 2235 2236 2237 2238 2239 2240 2241 2242 2243 2244 2245 2246 2247 2248 2249 2250 2251 2252 2253 2254 2255

Journal of Agricultural and Food Chemistry

(49) Berker, K. I.; Güçlü, K.; Demirata, B.; Apak, R. A novel antioxidant assay of ferric reducing capacity measurement using ferrozine as the colour forming complexation reagent. Anal. Methods 2010, 2, 1770-1778. (50) Işık, E.; Şahin, S.; Demir, C. Development of a new chromium reducing antioxidant capacity (CHROMAC) assay for plants and fruits. Talanta 2013, 111, 119-124. (51) Özyurt, D.; Demirata, B.; Apak, R. Determination of total antioxidant capacity by a new spectrophotometric method based on Ce(IV) reducing capacity measurement. Talanta 2007, 71, 1155-1165. (52) Özyurt, D.; Demirata Öztürk, B.; Apak, R. Modified cerium(IV)-based antioxidant capacity (CERAC) assay with selectivity over citric acid and simple sugars. J. Food Compos. Anal. 2010, 23, 282-288. (53) Özyurt, D.; Demirata, B.; Apak, R. Determination of total antioxidant capacity by a new spectrofluorometric method based on Ce(IV) reduction: Ce(III) fluorescence probe for CERAC assay. J. Fluoresc. 2011, 21, 2069-2076. (54) Scampicchio, M.; Wang, J.; Blasco, A. J.; Arribas, A. S.; Mannino, S.; Escarpa, A. Nanoparticle-based assays of antioxidant activity. Anal. Chem. 2006, 78, 2060-2063. (55) Özyürek, M.; Güngör, N.; Baki, S.; Güçlü, K.; Apak, R. Development of a silver nanoparticle-based method for the antioxidant capacity measurement of polyphenols. Anal. Chem. 2012, 84, 8052-8059. (56) Apak, R.; Güçlü, K.; Demirata, B.; Özyürek, M.; Çelik, S. E.; Bektaşoğlu, B.; Berker, K. I.; Özyurt, D. Comparative evaluation of total antioxidant capacity assays applied to phenolic compounds, and the CUPRAC assay. Molecules 2007, 12, 14961547. (57) Wolfenden, B. S.; Wilson, R. L. Radical-cations as reference chromogens in kinetic studies of one-electron transfer reactions: Pulse radiolysis studies of 2,2'azinobis-(3-ethylbenzthiazoline-6- sulphonate). J. Chem. Soc. Perkin Trans. 1982, 2, 805-812. (58) Niki, E. Antioxidant capacity: Which capacity and how to assess it? J. Berry Res. 2011, 1, 169-176. (59) Ou, B.; Huang, D.; Hampsch-Woodill, M.; Flanagan, J. A.; Deemer, E. K. Analysis of antioxidant activities of common vegetables employing oxygen radical absorbance capacity (ORAC) and ferric reducing antioxidant power (FRAP) assays: a comparative study. J. Agric. Food Chem. 2002, 50, 3122-3128. (60) Cao, G.; Prior, R. L. Comparison of different analytical methods for assessing total antioxidant capacity of human serum. Clin. Chem. 1998, 44, 1309-1315. (61) Prieto, P.; Pineda, M.; Aguilar, M. Spectrophotometric quantitation of antioxidant capacity through the formation of a phosphomolybdenum complex: specific application to the determination of vitamin E. Anal. Biochem. 1999, 269, 337-341. (62) Slinkard, K.; Singleton, V. L. Total phenol analysis: Automation and comparison with manual methods Am. J. Enol. Vitic. 1977, 28, 49-55. (63) Berker, K. I.; Olgun, F. A. Ö.; Özyurt, D.; Demirata, B.; Apak, R. Modified Folin−Ciocalteu antioxidant capacity assay for measuring lipophilic antioxidants J. Agric. Food Chem. 2013, 61, 4783-4791. (64) Demirci Çekiç, S.; Sözgen Başkan, K.; Tütem, E.; Apak, R. Modified cupric reducing antioxidant capacity (CUPRAC) assay for measuring the antioxidant capacities of thiol−containing proteins in admixture with polyphenols. Talanta 2009, 79, 344-351. 85 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

2256 2257 2258 2259 2260 2261 2262 2263 2264 2265 2266 2267 2268 2269 2270 2271 2272 2273 2274 2275 2276 2277 2278 2279 2280 2281 2282 2283 2284 2285 2286 2287 2288 2289 2290 2291 2292 2293 2294 2295 2296 2297 2298 2299 2300 2301 2302 2303 2304

Page 86 of 105

(65) Bean, H.; Radu, F.; De, E.; Schuler, C.; Leggett, R. E.; Levin, R. M. Comparative evaluation of antioxidant reactivity within obstructed and control rabbit urinary bladder tissue using FRAP and CUPRAC assays. Mol. Cell. Biochem. 2009, 323, 139-142. (66) Sözgen Başkan, K.; Tütem, E.; Özer, N.; Apak, R. Spectrophotometric and chromatographic assessment of contributions of carotenoids and chlorophylls to the total antioxidant capacities of plant foods. J. Agric. Food Chem. 2013, 61, 1137111381. (67) Karoui, H.; Hogg, N.; Frejaville, C.; Tordo, P.; Kalyanaraman, B. Characterization of sulfur-centered radical intermediates formed during the oxidation of thiols and sulfite by peroxynitrite. J. Biol. Chem. 1996, 271, 6000-6009. (68) Antolovich, M.; Prenzler, P. D.; Patsalides, E.; McDonald, S.; Robards, K. Methods for testing antioxidant activity. Analyst 2002, 127, 183-198. (69) Bors, W.; Heller, W.; Michel, C.; Saran, M. Flavonoids as antioxidants: Determination of radical scavenging efficiencies. Methods Enzymol. 1990, 186, 343355. (70) Rice-Evans, C. A.; Miller, N. J.; Paganga, G. Structure‒antioxidant activity relationships of flavonoids and phenolic acids. Free Radical Biol. Med. 1996, 20, 933-956. (71) Tütem, E.; Apak, R.; Baykut, F. Spectrophotometric determination of trace amounts of copper(I) and reducing agents with neocuproine in the presence of copper(II). Analyst 1991, 116, 89-94. (72) Lappin, A. G.; Youngblood, M. P.; Margerum, D. W. Electron-transfer reactions of copper(I) and copper(III) complexes. Inorg. Chem. 1980, 19, 407-413. (73) Bener, M.; Özyürek, M.; Güçlü, K.; Apak, R. Development of a low-cost optical sensor for cupric reducing antioxidant capacity measurement of food extracts. Anal. Chem. 2010, 82, 4252-4258. (74) Apak, R.; Güçlü, K.; Özyürek, M.; Çelik, S. E. Mechanism of antioxidant capacity assays and the CUPRAC (cupric ion reducing antioxidant capacity) assay. Microchim. Acta 2008, 160, 413-419. (75) Campos, C.; Guzmán, R.; López-Fernandez, E.; Casado, A. Evaluation of the copper(II) reduction assay using bathocuproinedisulfonic acid disodium salt for the total antioxidant capacity assessment: The CUPRAC–BCS assay. Anal. Biochem. 2009, 392, 37-44. (76) Smith, P. K.; Krohn, R. I.; Hermanson, G. T.; Mallia, A. K.; Gartner, F. H.; Provenzano, M. D.; Fujimoto, E. K.; Goeke, N. M.; Olson, B. J.; Klenk, D. C. Measurement of protein using bicinchoninic acid. Anal. Biochem. 1985, 150, 76-85. (77) Litwinienko, G.; Ingold, K. U. Solvent effects on the rates and mechanisms of reaction of phenols with free radicals. Acc. Chem. Res. 2007, 40, 222-230. (78) Çelik, S. E.; Özyürek, M., Güçlü, K.; Apak, R. Differences in responsivity of original cupric reducing antioxidant capacity and cupric-bathocuproine sulfonate assays to antioxidant compounds. Anal. Biochem. 2012, 423, 36-38. (79) Zhou, F.; Millhauser, G. L. The rich electrochemistry and redox reactions of the copper sites in the cellular prion protein. Coord. Chem. Rev. 2012, 256, 22852296. (80) Marques, S. S.; Magalhaes, L. M.; Tóth, I. V.; Segundo, M. A. Insights on antioxidant assays for biological samples based on the reduction of copper complexes‒the importance of analytical conditions. Int. J. Mol. Sci. 2014, 15, 11387-11402.

86 ACS Paragon Plus Environment

Page 87 of 105

2305 2306 2307 2308 2309 2310 2311 2312 2313 2314 2315 2316 2317 2318 2319 2320 2321 2322 2323 2324 2325 2326 2327 2328 2329 2330 2331 2332 2333 2334 2335 2336 2337 2338 2339 2340 2341 2342 2343 2344 2345 2346 2347 2348 2349 2350 2351 2352 2353

Journal of Agricultural and Food Chemistry

(81) Huang, X.; Cuajungco, M. P.; Atwood, C. S.; Hartshorn, M. A.; Tyndall, J. D. A.; Hanson, G. R.; Stokes, K. C.; Leopold, M.; Multhaup, G.; Goldstein, L. E.; Scarpa, R. C.; Saunders, A. J.; Lim, J.; Moir, R. D.; Glabe, C.; Bowden, E. F.; Masters, C. L.; Fairlie, D. P.; Tanzi, R. E.; Bush, A. I. Cu(II) potentiation of Alzheimer Aβ neurotoxicity: Correlation with cell-free hydrogen peroxide production and metal reduction. J. Biol. Chem. 1999, 274, 37111-37116. (82) Apak, R.; Güçlü, K.; Özyürek, M.; Bektaşoğlu, B.; Bener, M. CUPRAC (cupric ion reducing antioxidant capacity) assay for antioxidants in human serum, and for hydroxyl radical scavengers. In Advanced Protocols in Oxidative Stress II (Series: Methods in Molecular Biology), Armstrong, D.; Ed.; Humana PressSpringer: Gainesville, FL, USA 2010, Vol. 594, pp. 215-239. (83) Özyürek, M.; Güçlü, K.; Bektaşoğlu, B.; Apak, R. Spectrophotometric determination of ascorbic acid by the modified CUPRAC method with extractive separation of flavonoids-La(III) complexes. Anal. Chim. Acta 2007, 588, 88-95. (84) Özyürek, M.; Bektaşoğlu, B.; Güçlü, K.; Güngör, N.; Apak, R. Simultaneous total antioxidant capacity assay of lipophilic and hydrophilic antioxidants in the same acetone-water solution containing 2% methyl-beta-cyclodextrin using the Cupric Reducing Antioxidant Capacity (CUPRAC) method. Anal. Chim. Acta 2008, 630, 28-39. (85) Bektaşoğlu, B.; Çelik, S. E.; Özyürek, M.; Güçlü, K.; Apak, R. Novel hydroxyl radical scavenging antioxidant activity assay for water-soluble antioxidants using a modified CUPRAC method. Biochem. & Biophys. Res. Commun. 2006, 345, 11941200. (86) Özyürek, M.; Bektaşoğlu, B.; Güçlü, K.; Apak, R. Measurement of xanthine oxidase inhibition activity of phenolics and flavonoids with a modified cupric reducing antioxidant capacity (CUPRAC) method. Anal. Chim. Acta 2009, 636, 4250. (87) Özyürek, M.; Bektaşoğlu, B.; Güçlü, K.; Güngör, N.; Apak, R. A novel hydrogen peroxide scavenging assay of phenolics and flavonoids using cupric reducing antioxidant capacity (CUPRAC) methodology J. Food Compos. Anal. 2010, 23, 689-698. (88) Çelik, S. E.; Özyürek, M.; Güçlü, K.; Apak, R. Determination of antioxidants by a novel on-line HPLC-cupric reducing antioxidant capacity (CUPRAC) assay with post-column detection. Anal. Chim. Acta 2010, 674, 79-88. (89) Bekdeşer, B.; Özyürek, M.; Güçlü, K.; Apak, R. tert-Butylhydroquinone as a spectroscopic probe for the superoxide radical scavenging activity assay of biological samples. Anal. Chem. 2011, 83, 5652-5660. (90) Christodouleas, D. C.; Fotakis, C.; Papadopoulos, K.; Calokerinos, A. C. Evaluation of total reducing power of edible oils. Talanta 2014, 130, 233-240. (91) Walling, C. Kinetics of the decomposition of H2O2 catalyzed by ferric EDTA complexes. Proc. Natl. Acad Sci. USA 1975, 72, 140-142. (92) Ribeiro, J. P. N.; Magalhaes, L. M.; Reis, S.; Lima, J. L. F. C.; Segundo, M. A. High-throughput total cupric ion reducing antioxidant capacity of biological samples determined using flow injection analysis and microplate-based methods. Anal. Sci. 2011, 27, 483-488. (93) Gorinstein, S.; Leontowicz, M.; Leontowicz, H.; Najman, K.; Namiesnik, J.; Park, Y. S.; Jung, S. T.; Kang, S. G.; Trakhtenberg, S. Supplementation of garlic lowers lipids and increases antioxidant capacity in plasma of rats. Nutr. Res. 2006, 26, 362368.

87 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

2354 2355 2356 2357 2358 2359 2360 2361 2362 2363 2364 2365 2366 2367 2368 2369 2370 2371 2372 2373 2374 2375 2376 2377 2378 2379 2380 2381 2382 2383 2384 2385 2386 2387 2388 2389 2390 2391 2392 2393 2394 2395 2396 2397 2398 2399 2400 2401 2402 2403

Page 88 of 105

(94) Yen, G. C.; Duh, P. D. Antioxidative properties of methanolic extracts from peanut hulls. J. Am. Oil Chem. Soc. 1993, 70, 383-386. (95) Berker, K. I.; Demirata, B.; Apak, R. Determination of total antioxidant capacity of lipophilic and hydrophilic antioxidants in the same solution by using ferric– ferricyanide assay. Food Anal. Methods 2012, 5, 1150-1158. (96) Adcock, J. L.; Francis, P. S.; Barnett, N. W. Acidic potassium permanganate as a chemiluminescence reagent-A Review. Anal. Chim. Acta 2007, 601, 36-67. (97) Francis, P. S.; Costin, J. W.; Conlan, X. A.; Bellomarino, S. A.; Barnett, J. A.; Barnett, N. W. A rapid antioxidant assay based on acidic potassium permanganate chemiluminescence. Food Chem. 2010, 122, 926-929. (98) Cacig, S. I.; Szabo, M. I.; Lupea, A. X. D. Spectrophotometric method for the study of the antioxidant activity applied on Ziziphus jujoba and Hydrangea paniculata aqueous extracts. Proc. Nat. Sci, Matica Srpska Novi Sad. 2006, 110, 8793. (99) Popovic, B. M.; Štajner, D.; Slavko, K.; Sandra, B. Antioxidant capacity of cornelian cherry (Cornus mas L.) – Comparison between permanganate reducing antioxidant capacity and other antioxidant methods. Food Chem. 2012, 134, 734741. (100) Blasco, A. J.; Crevillen, A. G.; Gonzalez, M. C.; Escarpa, A. Direct electrochemical sensing and detection of natural antioxidants and antioxidant capacity in vitro systems. Electroanalysis 2007, 19, 2275-2286. (101) Hotta, H.; Sakamoto, H.; Nagano, S.; Osakai, T.; Tsujino, Y. Unusually large numbers of electrons for the oxidation of polyphenolic antioxidants. Biochim. Biophys. Acta 2001, 1526, 159-167. (102) Chevion, M.; Berenshtein, E.; Stadtman, E. R. Human studies related to protein oxidation: protein carbonyl content as a marker of damage. Free Radical Res. 2000, 33, 99-108. (103) Chevion, S.; Berry, E. M.; Kitrossky, N.; Kohen, R. Evaluation of plasma low molecular weight antioxidant capacity by cyclic voltammetry. Free Radical Biol. Med. 1997, 22, 411-421. (104) Kohen, R.; Vellaichamy, E.; Hrbac, J.; Gati, I.; Tirosh, O. Quantification of the overall reactive oxygen species scavenging capacity of biological fluids and tissues. Free Radical Biol. Med. 2000, 28, 871-879. (105) Piljac-Žegarac, J.; Valek, L.; Stipčević, T. T.; Martinez, S. Electrochemical determination of antioxidant capacity of fruit tea infusions. Food Chem. 2010, 121, 820-825. (106) Novak, I.; Šeruga, M.; Komorsky-Lovrić, Š. Electrochemical characterization of epigallocatechin gallate using square-wave voltammetry. Electroanal. 2009, 21, 1019-1025. (107) Magarelli, G.; Da Silva, J. G.; De Sousa Filho, I. A.; Dourado Lopes, I. S.; Rodrigues SouzaDe, J.; Hoffmann, L. V.; De Castro, C. S. P. Development and validation of a voltammetric method for determination of total phenolic acids in cotton cultivars. Microchem. J. 2013, 109, 23-28. (108) Korotkova, E. I.; Karbainov, Y. A.; Shevchuk, A.V. Study of antioxidant properties by voltammetry. J. Electroanal. Chem. 2002, 518, 56-60. (109) Kilmartin, P. A.; Zou, H.; Waterhouse, A. L. A cyclic voltammetry method suitable for characterizing antioxidant properties of wine and wine phenolics. J. Agric. Food Chem. 2001, 49, 1957-1965. (110) Campanella, L.; Bonanni, A.; Bellantoni, D.; Favero, G.; Tomassetti, M. Comparison of fluorimetric, voltammetric and biosensor methods for the 88 ACS Paragon Plus Environment

Page 89 of 105

2404 2405 2406 2407 2408 2409 2410 2411 2412 2413 2414 2415 2416 2417 2418 2419 2420 2421 2422 2423 2424 2425 2426 2427 2428 2429 2430 2431 2432 2433 2434 2435 2436 2437 2438 2439 2440 2441 2442 2443 2444 2445 2446 2447 2448 2449 2450 2451

Journal of Agricultural and Food Chemistry

determination of total antioxidant capacity of drug products containing acetylsalicylic acid. J. Pharm. Biomed. Anal. 2004, 36, 91-99. (111) Milardović, S.; Ivekovic, D.; Grabarić, B. S. A novel amperometric method for antioxidant activity determination using DPPH free radical. Bioelectrochem. 2006, 68, 175-180. (112) Gulaboski, R.; Mirčeski, V.; Mitrev, S. Development of a rapid and simple voltammetric method to determine total antioxidative capacity of edible oils. Food Chem. 2013, 138, 116-121. (113) Ferreira, R. Q.; Avaca, L. A. Electrochemical determination of the antioxidant capacity: The ceric reducing/antioxidant capacity (CRAC) assay. Electroanal. 2008, 20, 1323-1329. (114) Tufan, A. N.; Baki, S.; Güçlü, K.; Özyürek, M.; Apak, R. A novel differential pulse voltammetric (dpv) method for measuring the antioxidant capacity of polyphenols-reducing cupric neocuproine complex. J. Agric. Food Chem. 2014, 62, 7111-7117. (115) Prieto-Simon, B.; Cortina, M.; Campas, M.; Calas-Blanchard, C. Electrochemical biosensors as a tool for antioxidant capacity assessment. Sensors Actuat. B 2008, 129, 459-466. (116) Tammeveski, K.; Tenno, T. T.; Mashirin, A. A.; Hillhouse, E. W.; Manning, P.; McNeil, C. J. Superoxide electrode based on covalently immobilized cyt c: modelling studies. Free Radical Biol. Med. 1998, 25, 973-978. (117) Ignatov, S.; Shishniashvili, D.; Ge, B.; Scheller, F. W.; Lisdat, F. Amperometric biosensor based on a functionalized gold electrode for the detection of antioxidants. Biosens. Bioelectron. 2002, 17, 191-199. (118) Tian, Y.; Mao, L.; Okajima, T.; Ohsaka, T. A carbon fiber microelectrodebased third-generation biosensor for superoxide anion. Biosens. Bioelectron. 2005, 21, 557-564. (119) Lvovich, V.; Scheeline, A. Amperometric sensors for simultaneous superoxide and hydrogen peroxide detection. Anal. Chem. 1997, 69, 454-462. (120) Liu, J.; Su, B.; Lagger, G.; Tacchini, P.; Girault, H. H. Antioxidant redox sensors based on DNA modified carbon screen-printed electrodes. Anal. Chem. 2006, 78, 6879-6886. (121) Gorjanovic, S. Z.; Novakovic, M. M.; Potkonjak, N. I.; Sužnjevic, D. Z. Antioxidant activity of wines determined by a polarographic assay based on hydrogen peroxide scavenge. J. Agric. Food Chem. 2010, 58, 4626-4631. (122) Gorjanović, S.; Komes, D.; Pastor, F. T.; Belscak-Cvitanović, A.; Pezo, L.; Hečimović, I.; Sužnjevic, D. Antioxidant capacity of teas and herbal infusions: polarographic assessment. J. Agric. Food Chem. 2012, 60, 9573-9580. (123) Sužnjević, D.; Petrović, M.; Pastor, F. T.; Veljović, M.; Zlatanović, S.; Antić, M.; Gorjanović, S. Reduction of Hg2+ by individual phenolics and complex samples and its application in polarographic antioxidant assay. J. Electrochem. Soc. 2015, 162, H428-H433. (124) Jensen, T. R.; Duval Malinsky, M.; Haynes, C. L.; Van Duyne, R. P. Nanosphere lithography: Tunable localized surface plasmon resonance spectra of silver nanoparticles. J. Phys. Chem. B 2000, 104, 10549-10556. (125) Schafer, F. Q.; Buettner, G. R. Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Radical Biol. Med. 2001, 30, 1191-1212.

89 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

2452 2453 2454 2455 2456 2457 2458 2459 2460 2461 2462 2463 2464 2465 2466 2467 2468 2469 2470 2471 2472 2473 2474 2475 2476 2477 2478 2479 2480 2481 2482 2483 2484 2485 2486 2487 2488 2489 2490 2491 2492 2493 2494 2495 2496 2497 2498 2499 2500

Page 90 of 105

(126) Güçlü, K.; Özyürek, M.; Güngör, N.; Baki, S.; Apak, R. Selective optical sensing of biothiols with Ellman’s reagent: 5,5’-dithio-bis(2-nitrobenzoic acid)-modified gold nanoparticles. Anal. Chim. Acta 2013, 794, 90-98. (127) Bain, C. D.; Biebuyck, H. A.; Whitesides, G. M. Comparison of self-assembled monolayers on gold: Coadsorption of thiols and disulfides. Langmuir 1989, 5, 723727. (128) Chen, S. J.; Chang H. T. Nile red-adsorbed gold nanoparticles for selective determination of thiols based on energy transfer and aggregation. Anal. Chem. 2004, 76, 3727-3734. (129) Li, H.; Ma, X.; Dong, J.; Qian, W. Development of methodology based on the formation process of gold nanoshells for detecting hydrogen peroxide scavenging activity. Anal. Chem. 2009, 81, 8916-8922. (130) Ma, X.; Li, H.; Dong, J.; Qian, W. Determination of hydrogen peroxide scavenging activity of phenolic acids by employing gold nanoshells precursor composites as nanoprobes. Food Chem. 2011, 126, 698-704. (131) Apak, R.; Gorinstein, S.; Böhm, V.; Schaich, K. M.; Özyürek, M.; Güçlü, K. Methods of measurement and evaluation of natural antioxidant capacity/activity. Pure Appl. Chem. 2013, 85, 957-998. (132) Tian, X.; Schaich, K. M. Effects of molecular structure on kinetics and dynamics of the trolox equivalent antioxidant capacity assay with ABTS+•. J. Agric. Food Chem. 2013, 61, 5511-5519. (133) Xie, J.; Schaich, K. M. Re-evaluation of the 2,2-diphenyl-1-picrylhydrazyl free radical (DPPH) asay for antioxidant activity. J. Agric. Food Chem. 2014, 62, 42514260. (134) Schaich, K. M.; Tian, X.; Xie, J. Hurdles and pitfalls in measuring antioxidant efficacy: A critical evaluation of ABTS, DDPH, and ORAC assays, J. Funct. Foods 2015, 14, 111-125. (135) Papariello, G. J.; Janish, M. A. M. Diphenylpicrylhydrazyl as an organic analytical reagent in the spectrophotometric analysis of phenols. Anal. Chem. 1966, 38, 211-214. (136) Brand-Williams, W.; Cuvelier, M. E.; Berset, C. Use of a free radical method to evaluate antioxidant activity. LWT-Food Sci. Technol. 1995, 28, 25-30. (137) Gomez-Alonso, S.; Fregapane, G.; Salvador, M. D.; Gordon, M. H. Changes in phenolic composition and antioxidant activity of virgin olive oil during frying. J. Agric. Food Chem. 2003, 51, 667-672. (138) Lebeau, J.; Furman, C.; Bernier, J. L.; Duriez, P.; Teissier, E.; Cotelle, N. Antioxidant properties of di-tert-butylhydroxylated flavonoids. Free Radical Biol. Med. 2000, 29, 900-912. (139) McGowan, J. C.; Powell, T.; Raw, R. The rates of reaction of α,α-diphenyl-βpicrylhydrazyl with certain amines and phenols. J. Chem. Soc. 1959, 1959, 31033110. (140) Hogg, J. S.; Lohmann, D. H.; Russell, K. S. The kinetics of reaction of 2,2diphenyl-1-picrylhydrazyl with phenols. Can. J. Chem. 1961, 39, 1588-1594. (141) Beltran-Orozco, M. C.; Oliva-Coba, T. G.; Gallardo-Velazquez, T. ; OsorioRevilla, G. Ascorbic acid, phenolic content, and antioxidant capacity of red, cherry, yellow and white types of pitaya cactus fruit (Stenocereus stellatus riccobono). Agrociencia 2009, 43, 153-161. (142) Corral-Aguayo, R. D.; Yahia, E. M.; Carrillo-Lopez, A.; Gonzalez-Aguilar, G. Correlation between some nutritional components and the total antioxidant capacity

90 ACS Paragon Plus Environment

Page 91 of 105

2501 2502 2503 2504 2505 2506 2507 2508 2509 2510 2511 2512 2513 2514 2515 2516 2517 2518 2519 2520 2521 2522 2523 2524 2525 2526 2527 2528 2529 2530 2531 2532 2533 2534 2535 2536 2537 2538 2539 2540 2541 2542 2543 2544 2545 2546 2547 2548

Journal of Agricultural and Food Chemistry

measured with six different assays in eight horticultural crops. J. Agric. Food Chem. 2008, 56, 10498-10504. (143) Fogliano, V.; Verde, V.; Randazzo, G. Method for measuring antioxidant activity and its application to monitoring the antioxidant capacity of wines. J. Agric. Food Chem. 1999, 47, 1035-1040. (144) Ak, T.; Gülçin, I. Antioxidant and radical scavenging properties of curcumin. Chem.-Biol. Interact. 2008, 174, 27-37. (145) Mehdi, M. M.; Rizvi, S. I. N,N-Dimethyl-p-phenylenediamine dihydrochloride-based method for the measurement of plasma oxidative capacity during human aging. Anal. Biochem. 2013, 436, 165-167. (146) Schöttker, B.; Saum, K.-U.; Jansen, E. H. J. M.; Boffetta, P.; Trichopoulou, A.; Holleczek, B.; Dieffenbach, A. K.; Brenner, H. Oxidative stress markers and allcause mortality at older age: A population-based cohort study, J. Gerontol. A Biol. Sci. Med. Sci. 2015, 70, 518-524. (147) Demirci Çekiç, S.; Avan, A. N.; Uzunboy, S.; Apak, R. A colourimetric sensor for the simultaneous determination of oxidative status and antioxidant activity on the same membrane: N,N-Dimethyl-p-phenylene diamine (DMPD) on Nafion, Anal. Chim. Acta 2015, 865, 60-70. (148) Schwarz, K.; Bertelsen, G.; Nissen, L. R.; Gardner, P. T.; Heinonen, M. I.; Hopia, A.; Huynh-Ba, T.; Lambelet, P.; McPhail, D.; Skibsted, L. H.; Tijburg, L., Investigation of plant extracts for the protection of processed foods against lipid oxidation. Comparison of antioxidant assays based on radical scavenging, lipid oxidation and analysis of the principal antioxidant compounds, Eur. Food Res. Technol. 2001, 212, 319-328. (149) Nagaoka, S.-I.; Nagai, K.; Fujii, Y.; Ouchi, A.; Mukai, K. Development of a new free radical absorption capacity assay method for antioxidants: Aroxyl radical absorption capacity (ARAC), J. Agric. Food Chem. 2013, 61, 10054-10062. (150) Wiseman, H.; Halliwell, B. Damage to DNA by reactive oxygen and nitrogen species: Role in inflammatory disease and progression to cancer. Biochem. J. 1996, 313, 17-29. (151) Bourdon, E.; Blache, D. The importance of proteins in defense against oxidation. Antioxid. Redox Signal. 2001, 3, 293-311. (152) Beers, R. F.; Sizer, I. W. A spectrophotometric method for measuring the breakdown of hydrogen peroxide by catalase. J. Biol. Chem. 1952, 195, 133-140. (153) Aruoma, O. I.; Murcia, A.; Butler, J.; Halliwell, B. Evaluation of the antioxidant and prooxidant actions of gallic acid and its derivatives. J. Agric. Food Chem. 1993, 41, 1880-1885. (154) Ching, T. L.; de Jong, J.; Bast, A. A Method for screening hypochlorous acid scavengers by inhibition of the oxidation of 5-thio-2-nitrobenzoic acid: Application anti-asthmatic drugs. Anal. Biochem. 1994, 218, 377-381. (155) Bartosz, G. Use of spectroscopic probes for detection of reactive oxygen species. Clin. Chim. Acta. 2006, 368, 53-76. (156) Wolfe, K. L.; Liu, R. H. Cellular antioxidant activity (CAA) assay for assessing antioxidants, foods, and dietary supplements. J. Agric. Food Chem. 2007, 55, 8896-8907. (157) López-Alarcóna, C.; Denicola, A. Evaluating the antioxidant capacity of natural products: A review on chemical and cellular-based assays. Anal. Chim. Acta. 2013, 763, 1-10.

91 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

2549 2550 2551 2552 2553 2554 2555 2556 2557 2558 2559 2560 2561 2562 2563 2564 2565 2566 2567 2568 2569 2570 2571 2572 2573 2574 2575 2576 2577 2578 2579 2580 2581 2582 2583 2584 2585 2586 2587 2588 2589 2590 2591 2592 2593 2594 2595 2596 2597

Page 92 of 105

(158) Wolfe, K. L.; Liu, R. H. Structure-activity relationship of flavonoids in the cellular antioxidant activity (CAA) assay. J. Agric. Food Chem. 2008, 56, 84048411. (159) Wardman, P. Fluorescent and luminescent probes for measurement of oxidative and nitrosative species in cells and tissues: Progress, pitfalls, and prospects. Free Radical Biol. Med. 2007, 43, 995-1022. (160) Aruoma, O. I.; Halliwell, B.; Hoey, B. M.; Butler, J. The antioxidant action of N-acetylcysteine: its reaction with hydrogen peroxide, hydroxyl radical, superoxide, and hypochlorous acid. Free Radical Biol. Med. 1989, 6, 593-597. (161) Barclay, L. R. C. 1992 Syntex Award Lecture. Model biomembranes: quantitative studies of peroxidation, antioxidant action, partitioning, and oxidative stress. Can. J. Chem. 1993, 71, 1-16. (162) Buettner, G. R. The pecking order of free radicals and antioxidants: lipid peroxidation, α-tocopherol, and ascorbate. Arch. Biochem. Biophys. 1993, 300, 535543. (163) Pietta, P.-G. Flavonoids as antioxidants. J. Nat. Prod. 2000, 63, 1035-1042. (164) El-Magoli, S. B.; Karel, M.; Yong, S. Acceleration of lipid oxidation by volatile products of hydroperoxide decomposition. J. Food Biochem. 1979, 3, 111123. (165) Pinchuk, I.; Lichtenberg, D. The mechanism of action of antioxidants against lipoprotein peroxidation, evaluation based on kinetic experiments. Prog. Lipid Res. 2002, 41, 279-314. (166) Yıldogan Beker, B.; Bakır, T.; Sonmezoglu, I.; Imer, F.; Apak, R. Antioxidant protective effect of flavonoids on linoleic acid peroxidation induced by copper(II)/ascorbic acid system. Chem. Phys. Lipids. 2011, 164, 732-739. (167) Marcus, R. A. Transfer reactions in chemistry. Theory and experiment. Pure Appl. Chem. 1997, 69, 13-30. (168) Finotti, E.; Di Majo, D. Influence of solvents on the antioxidant property of flavonoids. Nahrung/Food. 2003, 47, 186-187. (169) Perez-Jimenez, J.; Saura-Calixto, F. Effect of solvent and certain food constituents on different antioxidant capacity assays. Food Res. Int. 2006, 39, 791800. (170) Pedrielli, P.; Pedulli, G. F.; Skibsted, L. H. Antioxidant mechanism of flavonoids. Solvent effect on rate constant for chain-breaking reaction of quercetin and epicatechin in autoxidation of methyl linoleate. J. Agric. Food Chem. 2001, 49, 3034-3040. (171) Pinelo, M.; Manzocco, L.; Nunez, M. J.; Nicoli, M. C. Solvent effect on quercetin antioxidant capacity. Food Chem. 2004, 88, 201-207. (172) Alfassi, Z. B.; Huie, R. E.; Neta, P. Substituent effects on rates of one-electron oxidation of phenols by the radicals ClO2, NO2, and SO3-. J. Phys. Chem. 1986, 90, 4156-4158. (173) Çelik, S. E.; Özyürek, M.; Güçlü, K.; Apak, R. Solvent effects on the antioxidant capacity of lipophilic and hydrophilic antioxidants measured by CUPRAC, ABTS/persulphate and FRAP methods. Talanta. 2010, 81, 1300-1309. (174) Choe, E.; Min, D. B. Mechanisms of antioxidants in the oxidation of foods, Comp. Rev. Food Sci. Food Safety 2009, 8, 345-357. (175) Foti, M.; Ruberto, G. Kinetic solvent effects on phenolic antioxidants determined by spectrophotometric measurements. J. Agric. Food Chem. 2001, 49, 342-348.

92 ACS Paragon Plus Environment

Page 93 of 105

2598 2599 2600 2601 2602 2603 2604 2605 2606 2607 2608 2609 2610 2611 2612 2613 2614 2615 2616 2617 2618 2619 2620 2621 2622 2623 2624 2625 2626 2627 2628 2629 2630 2631 2632 2633 2634 2635 2636 2637 2638 2639 2640 2641 2642 2643 2644 2645

Journal of Agricultural and Food Chemistry

(176) Barclay, L. R. C.; Edwards, C. E.; Vinqvist, M. R. Media effects on antioxidant activities of phenols and catechols. J. Am. Chem. Soc. 1999, 121, 6226-6231. (177) Leopoldini, M.; Marino, T.; Russo, N.; Toscano, M. Antioxidant properties of phenolic compounds: H-atom versus electron transfer mechanism. J. Phys. Chem. A. 2004, 108, 4916-4922. (178) Lucarini, M.; Mugnaini, V.; Pedulli, G. F. Bond dissociation enthalpies of polyphenols:  The importance of cooperative effects. J. Org. Chem. 2002, 67, 928931. (179) Barclay, L. R. C.; Vinqvist, M. R.; Mukai, K.; Goto, H.; Hashimoto, Y.; Tokunaga, A.; Uno, H. On the antioxidant mechanism of curcumin: classical methods are needed to determine antioxidant mechanism and activity. Org. Lett. 2000, 2, 2841-2843. (180) De Heer, M. I.; Mulder, P.; Korth, H.; Ingold, K. U.; Lusztyk, J. Hydrogen atom abstraction kinetics from intramolecularly hydrogen bonded ubiquinol-0 and other (poly)methoxy phenols. J. Am. Chem. Soc. 2000, 122, 2355-2360. (181) Wayner, D. D.; Lusztyk, E.; Page, D.; Ingold, K. U.; Mulder, P.; Laarhoven, L. J. J.; Aldrichs, H. S. Effects of solvation on the enthalpies of reaction of tert-butoxyl radicals with phenol and on the calculated O-H bond strength in phenol. J. Am. Chem. Soc. 1995, 117, 8738-8744. (182) Litwinienko, G.; Ingold, K. U. Abnormal effects on hydrogen atom abstractions. 1. The reactions of phenols with 2,2-diphenyl-1-picrylhydrazyl (DPPH) in alcohols. J. Org. Chem. 2003, 68, 3433-3438. (183) Pannala, A. S.; Chan, T. S.; O’Brien, P. J.; Rice-Evans, C. A. Flavonoid B-ring chemistry and antioxidant activity: Fast reaction kinetics. Biochem. Biophys. Res. Commun. 2001, 282, 1161-1168. (184) McPhail, D. B.; Hartley, R. C.; Gardner, P. T.; Duthie, G. G. Kinetic and stoichiometric assessment of the antioxidant activity of flavonoids by electron spin resonance spectroscopy. J. Agric. Food Chem. 2003, 51, 1684-1690. (185) Mukai, K.; Oka, W.; Watanabe, K.; Egawa, Y.; Nagaoka, S. Kinetic study of free-radical-scavenging action of flavonoids in homogeneous and aqueous triton X100 micellar solutions. J. Phys. Chem. A. 1997, 101, 3746-3753. (186) Burton, G. W.; Ingold, K. U. Autoxidation of biological molecules. 1. Antioxidant activity of vitamin E and related chain-breaking phenolic antioxidants in vitro. J. Am. Chem. Soc. 1981, 103, 6472-6477. (187) Sichel, G.; Corsaro, C.; Scalia, M.; Di Bilio, A. J.; Bonomo, R. P. In vitro scavenger activity of some flavonoids and melanins against O2•-. Free Radical Biol. Med. 1991, 11, 1-8. (188) Shahidi, F.; Wanasundara, P. K. J. Phenolic antioxidants. Crit. Rev. Food Sci. Nutr. 1992, 32, 67-103. (189) Di Meo, F.; Lemaur, V.; Cornil, J.; Lazzaroni, R.; Duroux, J.-L.; Olivier, Y.; Trouillas, P. Free radical scavenging by natural polyphenols: Atom versus electron transfer, J. Phys. Chem. A 2013, 117, 2082-2092. (190) Warren, J. J.; Tronic, T. A.; Mayer, J. M. Thermochemistry of proton-coupled electron transfer reagents and its implications. Chem. Rev. 2010, 110, 6961-7001. (191) Amić, A.; Marković, Z.; Marković, J. M. D.; Stepanić, V.; Lucić, B.; Amić, D. Towards an improved prediction of the free radical scavenging potency of flavonoids: The significance of double PCET mechanisms. Food Chem. 2014, 152, 578-585.

93 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

2646 2647 2648 2649 2650 2651 2652 2653 2654 2655 2656 2657 2658 2659 2660 2661 2662 2663 2664 2665 2666 2667 2668 2669 2670 2671 2672 2673 2674 2675 2676 2677 2678 2679 2680 2681 2682 2683 2684 2685 2686 2687 2688 2689 2690 2691 2692 2693 2694 2695

Page 94 of 105

(192) Bakhouche, K.; Dhaouadi, Z.; Jaidane, N.; Hammoutène, D. Comparative antioxidant potency and solvent polarity effects on HAT mechanisms of tocopherols, Comput. Theoretic. Chem. 2015, 1060, 58-65. (193) Basolo, F.; Pearson, R. G. Mechanism of Inorganic Reactions-A Study of Metal Complexes in Solution; John Wiley & Sons: New York, 1960; pp. 303-331. (194) Laguerre, M.; Giraldo, L. J. L.; Lecomte, J.; Figueroa-Espinoza, M. C.; Barea, B.; Weiss, J.; Decker, E. A.; Villeneuve, P. Chain length affects antioxidant properties of chlorogenate esters in emulsion: The cutoff theory behind the polar paradox. J. Agric. Food Chem. 2009, 57, 11335-11342. (195) Laguerre, M.; Giraldo, L. J. L.; Lecomte, J.; Figueroa-Espinoza, M. C.; Barea, B.; Weiss, J.; Decker, E. A.; Villeneuve, P. Relationship between hydrophobicity and antioxidant ability of “phenolipids” in emulsion: A parabolic effect of the chain length of rosmarinate esters. J. Agric. Food Chem. 2010, 58, 2869-2876. (196) Lucas, R.; Comelles, F.; Alcántara, D.; Maldonado, O. S;, Curcuroze, M.; Parra, J. L.; Morales, J. C. Surface-active properties of lipophilic antioxidants tyrosol and hydroxytyrosol fatty acid esters: A potential explanation for the nonlinear hypothesis of the antioxidant activity in oil-in-water emulsions. J. Agric. Food Chem. 2010, 58, 8021-8026. (197) Shahidi, F.; Zhong, Y. Revisiting the polar paradox theory: A critical overview. J. Agric. Food Chem. 2011, 59, 3499-3504. (198) Panya, A. Strategies to Improve the Performance of Antioxidants in Oil-inWater Emulsions. Ph.D. Thesis, University of Massachusetts, 2012. (199) A. Mortensen, L. H. Skibsted, A. Willnow, S. A. Everett, Re-appraisal of the tocopheroxyl radical reaction with beta-carotene: evidence for oxidation of vitamin E by the beta-carotene radical cation, Free Radical Res. 28 (1998) 69-80. (200) Bisby, R. H.; Parker, A. W. Reaction of ascorbate with the α-tocopheroxyl radical in micellar and bilayer membrane systems. Arch. Biochem. Biophys. 1995, 317, 170-178. (201) Pedrielli, P.; Skibsted, L. H. Antioxidant synergy and regeneration effect of quercetin, (−)-epicatechin, and (+)-catechin on alpha-tocopherol in homogeneous solutions of peroxidating methyl linoleate. J. Agric. Food Chem. 2002, 50, 71387144. (202) Hudson, B. J. F.; Lewis, J. I. Polyhydroxy flavonoid anti-oxidant for edible oils-structural criteria for activity, Food Chem. 1983, 10, 147-155. (203) Peyrat-Maillard, M. N.; Cuvelier, M. E.; Berset, C. Antioxidant activity of phenolic compounds in 2,2’-azobis (2-amidinopropane) dihydrochloride (AAPH)induced oxidation: synergistic and antagonistic effect, J. Am. Oil Chem. Soc. 2003, 80, 1007-1012. (204) Hidalgo, M.; Sánchez-Moreno, C.; de Pascual-Teresa, S. Flavonoid-flavonoid interaction and its effect on their antioxidant activity, Food Chem. 2010, 121, 691696. (205) Wang, S.; Meckling, K. A.; Marcone, M. F.; Kakuda, Y.; Tsao, R. Synergistic, additive, and antagonistic effects of food mixtures on total antioxidant capacities, J. Agric. Food Chem. 2011, 59, 960-968. (206) Skroza, D.; Mekinić, I. G.; Svilović, S.; Simat, V.; Katalinić, V. Investigation of the potential synergistic effect of resveratrol with other phenolic compounds: A case of binary phenolic mixtures, J. Food Compos. Anal. 2015, 38, 13-18. (207) Prieto, M. A.; Curran, T. P.; Gowen, A.; Vázquez, J. A. An efficient methodology for quantification of synergy and antagonism in single electron transfer antioxidant assays, Food Res. Int. 2015, 67, 284-298. 94 ACS Paragon Plus Environment

Page 95 of 105

2696 2697 2698 2699 2700 2701 2702 2703 2704 2705 2706 2707 2708 2709 2710

Journal of Agricultural and Food Chemistry

(208) Vitaglione, P.; Napolitano, A.; Fogliano, V. Cereal dietary fibre: a natural functional ingredient to deliver phenolic compounds into the gut. Trends Food Sci. Technol. 2008, 19, 451-463. (209) Serpen, A.; Capuano, E.; Fogliano, V.; Gökmen, V. A new procedure to measure the antioxidant activity of insoluble food components. J. Agric. Food Chem. 2007, 55, 7676-7681. (210) Tufan, A. N.; Çelik S. E.; Özyürek, M.; Güçlü, K.; Apak, R. Direct measurement of total antioxidant capacity of cereals: QUENCHER-CUPRAC method. Talanta. 2013, 108, 136-142. (211) Serpen, A.; Gökmen, V.; Fogliano, V.; Solvent effects on total antioxidant capacity of foods measured by direct QUENCHER procedure, J. Food Compos. Anal. 2012, 26, 52-57. (212) Gökmen, V.; Serpen, A.; Fogliano, V. Direct measurement of the total antioxidant capacity of foods: The ‘QUENCHER’ approach. Trends Food Sci. Technol. 2009, 20, 278-288.

95 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 96 of 105

Figure Captions: Figure 1. Direct (competitive) antioxidant assay, involving a fluorogenic or chromogenic probe and biologically relevant ROS/RNS. Figure 2. Indirect (non-competitive) antioxidant assay, in which physiological redox reactions (i.e., oxidant-antioxidant interactions) are simulated on an artificial probe without biologically relevant ROS/RNS. Figure 3. Characteristic functional groups having a key role in high antioxidant capacity of a flavonoid (like quercetin). Figure 4. Reaction scheme for the CUPRAC antioxidant capacity assay (liberated protons are buffered by ammonium acetate). Figure 5. Methyl-β-cyclodextrin oligosaccharide, retaining lipophilic antioxidants in the hydrophobic inner core, while holding hydrophilic antioxidants on the outer surface. Figure 6. (a) Salicylate exposed to hydroxyl radicals generated in a Fenton system is converted to highly CUPRAC-reactive species (CUPRAC-rs), i.e. dihydroxy-benzoate (DHBA) isomers, enabling a turn-on colorimetric assay for hydroxyl radicals. (b) Primary hydroxylation products of salicylate. Figure 7. The superoxide anion radicals generated by a non-enzymatic PMS-NADH system attack the CUPRAC-reactive TBHQ probe, converting it to non-reactive TBB-quinone (TBBQ); this conversion is less in the presence of antioxidants, enabling a modified CUPRAC assay for superoxide scavenging antioxidants. Figure 8. Enlargement of silver nanoparticles by antioxidant addition to a silver nitrate colloidal solution of citrate-stabilized Ag-nanoparticles (via seed-mediated particle growth to generate core-shell AgNPs). Figure 9. Surface plasmon resonance absorption of citrate-stabilized AgNPs is intensified by the addition of apple juices, corresponding to seed-mediated particle growth. Figure 10: AuNPs−adsorbed Ellman’s reagent (DTNB-disulfide) is exchanged for thiols (SH) in solution, enabling a colorimetric thiol assay.126

96 ACS Paragon Plus Environment

Page 97 of 105

Journal of Agricultural and Food Chemistry

Figure 1

97 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Figure 2

98 ACS Paragon Plus Environment

Page 98 of 105

Page 99 of 105

Journal of Agricultural and Food Chemistry

Figure 3

Figure 4

99 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Figure 5

100 ACS Paragon Plus Environment

Page 100 of 105

Page 101 of 105

Journal of Agricultural and Food Chemistry

Figure 6

101 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Figure 7

102 ACS Paragon Plus Environment

Page 102 of 105

Page 103 of 105

Journal of Agricultural and Food Chemistry

Figure 8

103 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Figure 9

Figure 10

104 ACS Paragon Plus Environment

Page 104 of 105

Page 105 of 105

Journal of Agricultural and Food Chemistry

TOC Graphic

105 ACS Paragon Plus Environment