CCA-13536; No of Pages 16 Clinica Chimica Acta xxx (2014) xxx–xxx

Contents lists available at ScienceDirect

Clinica Chimica Acta journal homepage: www.elsevier.com/locate/clinchim

F

O

4 5Q2 6 7 8 9

a

1 0

a r t i c l e

11 12 13 14 15

Article history: Received 4 April 2014 Received in revised form 4 June 2014 Accepted 5 June 2014 Available online xxxx

16 17 18 19 20

Keywords: Antioxidant Oxidative stress Diabetes Cancer

NPO-International Laboratory of Biochemistry, 1-166, Uchide, Nakagawa-ku, Nagoya 454-0926, Japan Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Israel Oncology Department Soroka University Medical Center, Be'er-Sheva 84105, Israel d Department of Applied Zoology and Biotechnology, Vivekananda College (A Gurukula Institute of Life Training), Affiliated to Madurai Kamaraj University, Thiruvedakam West, Madurai 625234, India e Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel f Department of Clinical Biochemistry and Pharmacology, Soroka University Medical Center, Ben-Gurion University of the Negev, Be'er-Sheva 84105, Israel b c

i n f o

a b s t r a c t

Oxidative stress plays a pivotal role in the development of human diseases. Reactive oxygen species (ROS) that includes hydrogen peroxide, hyphochlorus acid, superoxide anion, singlet oxygen, lipid peroxides, hypochlorite and hydroxyl radical are involved in growth, differentiation, progression and death of the cell. They can react with membrane lipids, nucleic acids, proteins, enzymes and other small molecules. Low concentrations of ROS has an indispensable role in intracellular signalling and defence against pathogens, while, higher amounts of ROS play a role in number of human diseases, including arthritis, cancer, diabetes, atherosclerosis, ischemia, failures in immunity and endocrine functions. Antioxidants presumably act as safeguard against the accumulation of ROS and their elimination from the system. The aim of this review is to highlight advances in understanding of the ROS and also to summarize the detailed impact and involvement of antioxidants in selected human diseases. © 2014 Published by Elsevier B.V.

E

T

C

34 32 31

Introduction . . . . . . . . . . . . . . . . . Measuring oxidative stress . . . . . . . . . . 2.1. Pro-oxidants . . . . . . . . . . . . . 3. Potential antioxidant biomarkers . . . . . . . 3.1. Superoxide dismutase (SOD) . . . . . . 3.2. Catalase (CAT) . . . . . . . . . . . . 3.3. Glutathione peroxidase (GPx) . . . . . 3.4. Thiol index . . . . . . . . . . . . . . 3.5. Xanthine oxidase (XO) . . . . . . . . . 3.6. NADPH Oxidase . . . . . . . . . . . . 3.7. A biomarker of cardiovascular damage . 4. Mechanism of antioxidant action and nutrients . 5. Antioxidants vs human diseases . . . . . . . . 5.1. Antioxidant vs arthritis . . . . . . . . . 5.2. Anti oxidant vs cancer . . . . . . . . . 5.3. Antioxidant vs diabetes . . . . . . . . 5.4. Antioxidant vs arthrosclerosis . . . . . 5.5. Anti oxidant vs neurodegerative diseases 6. Conclusion . . . . . . . . . . . . . . . . . 7. Uncited references . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

N C O

R

R

1. 2.

U

37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57

Contents

E

33 36 35

R O

3

Peramaiyan Rajendran a, Natarajan Nandakumar b, Thamaraiselvan Rengarajan a, Rajendran Palaniswami d, Edwinoliver Nesamony Gnanadhas e, Uppalapati Lakshminarasaiah f, Jacob Gopas b,c, Ikuo Nishigaki b,⁎

P

2

Antioxidants and human diseases

D

1

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . .

58 ⁎ Corresponding author. E-mail address: [email protected] (I. Nishigaki).

http://dx.doi.org/10.1016/j.cca.2014.06.004 0009-8981/© 2014 Published by Elsevier B.V.

Please cite this article as: Rajendran P, et al, Antioxidants and human diseases, Clin Chim Acta (2014), http://dx.doi.org/10.1016/j.cca.2014.06.004

0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0

21 22 23 24 25 26 27 28 29 30

P. Rajendran et al. / Clinica Chimica Acta xxx (2014) xxx–xxx

94 95

98 99

As oxidative stress is indicative of an inequity between oxidants and antioxidants, methods for quantifying oxidative stress mostly include straight or indirect measurement of oxidants and antioxidants [6,7]. In the following segment, some principles and commonly used methods for the measurement of oxidative stress and damage will be briefly outlined.

100

2.1. Pro-oxidants

101 102

The most abundant free radicals in biological systems are the oxygen-centered free radicals and their metabolites, usually referred to as ROS [7]. ROS are formed continuously as normal by-products of cellular metabolism; and, in low concentrations, they are essential for several physiological processes, including protein phosphorylation, transcription factor activation, cell differentiation, apoptosis, oocyte maturation, steroidogenesis, cell immunity, and cellular defense against microorganisms [7]. However, when produced in excess, ROS can damage cell functionality as they can harm cellular lipids, proteins, and DNA [7,8]. The plasma level of ROMs is considered an indicator of free-radical production [7]. ROMs is a collective term that includes not only oxygencentered free radicals such as the superoxide anion and hydroxyl radical, but also some non-radical derivatives of oxygen, such as hydrogen peroxide (H2O2) and hypochlorous acid [9]. A ROMs kit has been developed to assess oxidant levels in plasma and other biological fluids; and the ROMs test has been validated by electron spin resonance [10], which is considered the “gold standard” for measuring total oxidative status. However, electron spin resonance is not suitable for routine analysis, as the method is complex and requires specific technical assistance

81 82 83 84 85 86 87 88 89 90

96 97

103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119

C

79 80

E

77 78

R

75 76

R

73 74

O

71 72

C

69 70

N

67 68

U

65 66

F

2. Measuring oxidative stress

64

O

93

62 63

R O

91 92

The beneficial effect of antioxidants on the maintenance of health in human has become an important subject that has engaged many scientists across the world over the last decade. In the last few years, antioxidants have become the indispensable nutrients of the nutritional world. Antioxidants are important in terms of their ability to protect against oxidative cell damage that can lead to conditions, such as Alzheimer's disease, cancer, heart disease and also linked with chronic inflammation [1]. It is defined as the substances which at low concentration significantly inhibits or delay the oxidative process, while often being oxidized themselves. Recent reports suggest that several endogenous and exogenous antioxidants are used to neutralize free radicals and protect the body from free radicals by maintaining redox balance [2,3]. Singh et al. (2010) [4] quoted that antioxidants have gone from “Miracle Molecules” to “Marvellous Molecules” and finally to “Physiological Molecules” that they play a vital role in metabolic pathways and protect cells. However recent conflicting evidence has forced the scientists to dig deeper in order to explore the role of antioxidants and pro-oxidants, since free radical reactions have been implicated in every human pathological condition which includes neurodegenerative disorders like Alzheimer's disease, Parkinson's disease, multiple sclerosis, amyotrophic lateral sclerosis, memory loss, depression and cardiovascular diseases such as atherosclerosis, ischemic heart disease, cardiac hypertrophy, hypertension, shock and trauma. Further, it also implicated in pulmonary disorders which include inflammatory lung diseases such as asthma and chronic obstructive pulmonary disease and additionally diseases associated with premature infants such as bronchopulmonary dysplasia, periventricular leukomalacia, intraventricular hemorrhage, retinopathy of prematurity and necrotizing enterocolitis and in some autoimmune diseases like rheumatoid arthritis and also in several renal disorders such as glomerulonephritis, tubulointerstitial nephritis, chronic renal failure, proteinuria, uremia and finally gastrointestinal diseases like peptic ulcer, inflammatory bowel disease and colitis, diabetes, tumors and cancers [5].

P

60 61

not available in most laboratories. The utility of the ROMs assay in monitoring oxidative stress in goats [11], sheep [12], and dairy cows [13] has been reported. The concentrations of individual oxidant components can be measured separately in the laboratory; but such measurements are time-consuming, labor intensive, and costly. Free-radical analytical system 4 technology has been shown to offer a quick, simple, precise, and reliable method for assessing the oxidative status in dairy cows [14] and in horses [15]. Such technology is particularly useful in the field, where it is not always practical or possible to get samples to a laboratory immediately. The possibility of assessing oxidative stress directly in blood samples provides veterinarians with a simple and reliable method for measuring oxidative stress in clinical situations such as the monitoring of therapy and in the antioxidant supplementation of domestic animals. However, given the lack of reference values for ROMs in ruminants, it is difficult to establish if and when these animals are actually experiencing oxidative stress. Therefore, it is important to calculate the specific referral ranges; because a correct biochemical evaluation of oxidative status is an essential premise to prevent and eventually to treat the effects of oxidative stress in ruminant medicine. Advanced oxidation protein products (AOPPs) are terminal products of proteins exposed to free radicals and arise from the reaction between plasma proteins and chlorinated oxidants mediated by myeloperoxidase, a neutrophil enzyme [16,17]. In humans, AOPPs have been linked to several diseases, such as chronic renal failure [18], diabetes mellitus [19], diabetic nephropathy [20], coronary artery diseases [21], and obesity [22]. Chronic accumulation of AOPPs has been demonstrated to promote inflammatory processes in the diabetic kidney [20] and in chronic renal failure [18], indicating that these products might be a byproduct of neutrophil activation during infections. Studies in ruminants have shown higher levels of AOPPs than normal in lambs [23] and dairy cows [24] supplemented with Yerba Mate (Ilex paraguanensis). More information about the role of protein oxidation in ruminants' health and about oxidated proteins could be obtained by a comparison of AOPPs with other indicators of protein oxidation, such as advanced glycation end products (AGE). However, a correlation between AGE and inflammatory parameters is usually not found or is only weak, and the induction of proinflammatory activities caused by AOPPs seems to be more intense [25,26], suggesting that oxidative stress is more closely linked to inflammation and acute-phase reactions than to the advanced glycation process and its end products. AOPPs could thus better describe acute inflammation, whereas AGE might serve more as a marker of chronic long-lasting damage [25]. These observations are highly relevant, as increased levels of AOPPs could indicate the presence of inflammatory processes that can potentially compromise the correct embryonic development in dairy cows [14,27]. Lipids, in particular those that are polyunsaturated, are prone to oxidation. Lipids are one of the most susceptible substrates to damage by free radicals, and biomarkers of lipid peroxidation are considered the best indicators of oxidative stress [28]. Malondialdehyde (MDA) is one of several low-molecular-weight end products formed during radical-induced decomposition of polyunsaturated fatty acids [29]. MDA readily reacts with thiobarbituric acid, producing a red pigment that can be easily measured by spectrophotometry in the form of thiobarbituric acid-reactive substances (TBARS, [29]). It is worth noting that the MDA assay has been criticized for its low specificity and for artifact formation, since only a fraction of the MDA measured is actually generated in vivo. Furthermore, the TBARS assay, a common method used to measure MDA, is considered inaccurate, and returns results that differ according to the assay conditions used [30]. For example, studies on dairy cows have yielded contrasting results, with some reports failing to show any significant changes in plasma MDA concentrations during the peripartum period [31]; whereas in other studies MDA or TBARS concentrations were shown to increase around calving [8,16,32]. This apparent discrepancy could also have been mainly due to the great variation in individual MDA concentrations measured in the studies by Castillo et al. [31]. Similarly, studies on

D

1. Introduction

T

59

E

2

Please cite this article as: Rajendran P, et al, Antioxidants and human diseases, Clin Chim Acta (2014), http://dx.doi.org/10.1016/j.cca.2014.06.004

120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185

P. Rajendran et al. / Clinica Chimica Acta xxx (2014) xxx–xxx

3.1. Superoxide dismutase (SOD)

211

216 217

The term superoxide dismutase characterizes a family of enzymatic proteins differing in their structure and cofactors, among them being Mn-SOD and Cu–Zn-SOD. SOD activity enhances the spontaneous dismutation of superoxide radicals to H2O2 [52]. SOD can be measured by utilizing the technique of Misra and Fridovich [43], which is based on inhibition of the formation of nicotine amide adenine dinucleotide, phenazine methosulfate, and amino blue tetrazolium formazan.

218

3.2. Catalase (CAT)

219 220

The end product of the dismutation reaction, i.e., H2O2, can be removed by the activity of the enzyme catalase. CAT is an enzyme with a very high KM for its substrate and can remove H2O2 present in high concentrations [52]. The activity of CAT can be measured through the

214 215

221 222

t1:1 t1:2

C

212 213

E

207 208

R

205 206

R

203 204

N C O

201 202

Table 1 Biomarkers of oxidative stress. Biomarkers

t1:4 t1:5 t1:6 t1:7 t1:8 t1:9 t1:10 t1:11 t1:12 t1:13 t1:14 t1:15 t1:16 t1:17 t1:18 t1:19 t1:20 t1:21 t1:22 t1:23 t1:24

Carbonyls Lipid peroxidation •Malondialdehyde •F2-isoprostane •4-Hydroxynonenal Ferric reducing ability of plasma Plasma vitamins •Vitamin C •Vitamin E Antioxidant enzymes •Superoxide dismutase •Catalase •Glutathione peroxidase •GSH/GSSG ratio in erythrocyte Prooxidant enzymes •Xanthine oxidase •NADPH oxidase Others •Endothelial microparticles •Endothelial progenitor cells •Ischemia modified albumin

U

t1:3

Location

Reference

Extracellular

Mohanty et al. [36]

Extracellular Extracellular Extracellular Extracellular

Ohkawa et al. [37] Collins et al. [38] Halliwell and Gutteridge [39] Benzie and Strain [40]

Extracellular Extracellular

Roe and Kuether [41] Teissier [42]

Intracellular Intracellular Intracellular Intracellular

Misra and Fridovich [43] Aebi and Bergmeyer [44] Floh'e andG¨unzler [45] Hissin and Hilf [46]

Intracellular Intracellular

Haining and Legan [47] Nauseef [48]

Extracellular Extracellular Extracellular

Burger and Touyz [49] Touyz and Schiffrin [50] Sinha et al. [51]

226 227

3.4. Thiol index

232

F

210

199 200

The biochemical function of glutathione peroxidase is to reduce lipid hydroperoxides to their corresponding alcohols and to reduce free H2O2 to water. In contrast to catalase, peroxidase possesses a high affinity for and can remove H2O2 even when it is present in a low concentration [52]. Its activity can be estimated by the method described by Flohe and Gunzler [45].

228 229 230 231

The GSH/GSSG index is a parameter of the intracellular redox status. GSH is the major endogenous antioxidant produced by cells; and it participates directly in the neutralization of free radicals and reactiveoxygen compounds, as well as in maintaining exogenous antioxidants such as vitamins C and E in their reduced forms [53]. GSH and GSSG levels can be measured in erythrocytes by the method described by Hissin and Hilf [46].

233

3.5. Xanthine oxidase (XO)

240

O

209

Epidemiologic literature that focuses on antioxidant status and chronic disease risk has in the past relied primarily upon biomarkers of exposure to antioxidant nutrients. However, this approach is in essence using exposure estimates to a select group of nutrients as a surrogate for estimating actual oxidative defense or oxidative stress status. Emphasis is now being placed on developing functional biomarkers of oxidative stress status, that is, biomarkers that integrate the effect of exposure to oxidants coupled with the full range of antioxidant protectivemechanisms in vivo. Many of such biomarkers are being studied including various measures of oxidation products of lipid, DNA, and protein (Table 1). Some of these biomarkers are now being applied in research of pathologies related to oxidative stress.

193 194

225

R O

198

192

3.3. Glutathione peroxidase (GPx)

P

3. Potential antioxidant biomarkers

190 191

234 235 236 237 238 239

XO is a key enzyme involved in the formation of reactive oxygen species, and it plays a major role in cell oxidative stress. This enzyme catalyzes the oxidation of hypoxanthine to xanthine and can further catalyze the oxidation of xanthine to uric acid [54]. Xanthine oxidase can be estimated by the method of Haining and Legan [47].

241

3.6. NADPH Oxidase

246

The NADPH-oxidase complex utilizes electrons to produce superoxide radicals from the oxygen molecule. NADPH oxidase may be measured by a chemiluminescence [55] or electrochemical method among others. Approaches to quantitate oxygen consumption, extracellular release of O∙2 or H2O2, and intracellular O∙2 production provide reliable assessment of NADPH oxidase activity in a given population of cells [56].

247

3.7. A biomarker of cardiovascular damage

253

Endothelial microparticles (EMPs) are nuclear fragments of cellular membrane shed from oxidatively stressed or damaged cells. Typically defined as having a diameter of 0.1 to 1.0 mm, these microparticles contain surface proteins and cytoplasmic material of the parental cells [57]. Many biomonitoring studies have investigated the role of antioxidants in reducing oxidatively generated DNA damage in urine and white blood cells. A collective interpretation is difficult because many studies lack sufficient control and have unreasonably high baseline levels of oxidatively damaged DNA. to investigating antioxidant effects has been the use of biomarkers of oxidatively damaged DNA. This is based on the mechanistic rationale that dietary antioxidants inhibit the oxidation of DNA. With the possibility of detecting 8-oxo-7,8dihydro-2′-deoxyguanosine (8-oxodG), the first of many antioxidant supplementation studies with focus on this lesion in WBC appeared in the beginning of the 1990s. At the same time, reliable detection of urinary 8-oxodG excretion was achieved, and this was soon followed by antioxidant supplementation studies using urinary 8-oxodG excretion as key biomarker. However, by far the most popular method in antioxidant intervention trials has been the comet assay that detects DNA strand breaks (SB). An enzyme-modified version of the comet assay has been developed to detect oxidatively altered nucleobases by including a DNA digestion step with DNA glycosylase or endonuclease enzymes [58]. The characteristics of the intervention studies are outlined in Table 2.

254

D

197

188 189

non spontaneous decomposition of hydrogen peroxide, by the method 223 described by Aebi [44]. 224

T

195 196

transported cattle have failed to report a consistent change in their MDA concentration. It seems that this discrepancy is partly due to the different methodologies employed to assess MDA levels. TBARS represent a wide range of lipid peroxidation products, and the thiobarbituric acid reaction is rather non-specific for MDA [33]. High-pressure liquid chromatography would be expected to be highly specific, and perhaps, more accurate than the spectrophotometric procedures [34]. More recently, an ELISAbased method for the detection of isoprostane, which is considered to be the most reliable marker of lipid oxidation [35], has become commercially available and might be able to shed more light on the role of lipid peroxidation during the peripartum period in ruminants.

E

186 187

3

Please cite this article as: Rajendran P, et al, Antioxidants and human diseases, Clin Chim Acta (2014), http://dx.doi.org/10.1016/j.cca.2014.06.004

242 243 244 245

248 249 250 251 252

255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277

4 t2:1 t2:2

P. Rajendran et al. / Clinica Chimica Acta xxx (2014) xxx–xxx

Table 2 Administrations of dietary antioxidant with assessment of oxidatively DNA damage in white blood cells and urine.

t2:3

Supplement per day

Subjects

Age (years)

Effect

References

t2:4 t2:5 t2:6

β-carotene (20 mg) for 14 weeks β-carotene (30 mg) for 1 months Carotenoidsa for 3 weeks

122 M (S) 14 M (NS) 32 MF(NS)

39 (NR) 19–22 32 ± 11

[59] [60] [61]

t2:7

Vitamin C (500 mg as plain or slow release formulations) and vitamin E (182 mg) for 4 weeks Multi-vitamin tablet (100 mg vitamin C, 280 mg vitamin E, and 25 mg b-carotene) for 20 weeks Vegetable consumption (3.6 vs 12 servings) for 2 weeks Kiwi fruit (1–3 pieces) for 3 weeks Cranberry juice (750 ml) for 2 weeks Blackcurrant juice or anthocyanine drink (475–1000 ml) for 3 weeks Green tea or black tea (four cups) for 1–4 months

48 M (S)

39 ± 12

No effect on urinary excretion of 8-oxodG/24-h (HPLC) No effect on urinary excretion of 8-oxodG/24-h (HPLC) No effect versus baseline, but decreased urinary excretion of 8-oxodG ELISA) in active group post-supplementation Decreased ENDOIII and FPG sites (Comet) in the group that ingested tablets with the slowrelease vitamin C formulation 100 M (50S) 50–59 Decreased ENDOIII sites (Comet) after 20 weeks in WBC

Two interventions of green tea with 300 ml for 7 days or 32 oz for 7 days Green tea extract b for 3 weeks

68 MF (13S) 18–45 16 M (8S)

20–31

124 MF(NR) 38 F (NR)

NR

t2:17

Green tea (500 or 1000 mg) among high-risk subjects of liver cancer for 3 months Soya-hypocotyl tea (N1000 ml) for 1 months

t2:18 t2:19

Soy milk, rice milk, or cow milk (1 l) for 4 weeks Polyphenol-rich olive oils (25 ml) for 40 days

10 M (NS) 12 M (NS)

20–50 20–22

t2:20

Olive oil (25 ml) with three different content of phenolic 182 M (NS) compounds for 3 weeks Antioxidant rich diet or tabletsc for 5 weeks 55 MF (NR)

t2:15 t2:16

t2:21

14 MF (NS) 20 F (2S) 57 MF (6S)

26–54 18–40 19–52

Decreased ENDOIII and FPG (Comet) sites No effect on ENDOIII (Comet) in WBC No effect on ENDOIII and FPG sites (Comet) in WBC

[65] [66] [67]

120 MF(S)

18–79

Decreased excretion of 8-oxodG (ELISA) in spot urine samples after 4 mo in green tea group, but not before Decreased 12-h urinary excretion of 8-oxodG (HPLC)

NR

20–60 71 ± 6

F

[64]

O

t2:14

Lower 8-oxodG in WBC (HPLC) among those subjects eating 12 servings per day

R O

t2:13

23–81

No effect related to supplementation on 24-h urinary excretion of 8-oxodG (HPLC) but a decreased excretion during the study in all groups Lower 8-oxodG/24-h (HPLC) after 3 months supplementation but not after 1 month Decreased excretion of 8-oxodG (ELISA) in active group (statistical test not reported) Decreased ENDOIII sites (Comet) for soy milk Decreased excretion of 8-oxodG (HPLC) in spot urinary samples following supplementation in a dose-dependent manner Decreased 8-oxodG/24-h urinary excretion, but no difference in urinary excretion of 8-oxoGua/24-h. No difference in the effect of the different olive oils No effect versus baseline, but decreased 24-h urinary excretion of 8-oxodG (ELISA) in active group post-supplementation

P

t2:10 t2:11 t2:12

64 F (NR)

D

t2:9

[63]

[68,69] [70] [71] [72] [73] [74] [75] [76] [77]

E

t2:8

[62]

Number of subjects indicated as males (M) and females (F). Smokers (S) and non-smokers (NS) are indicated in brackets. Lacking information is indicated as NR (not reported) Age is shown as range or mean ± standard deviation. aSupplement constitute b-carotene (6 mg), a-carotene (1.4 mg), lycopene (4.5 mg), bixin (11.7 mg), lutein (4.4 mg), and paprika carotenoids (2.2 mg), b Consisted of 1000 mg extract/kg bodyweight in meat patties (total phenolics were 23.5 mg/10 MJ), cConsisted of tablets with vitamin antioxidants (400 mg vitamin C, 150 mg vitamin E, 4 mg b-carotene), capsules (90 mg vitamin C, 18 IU vitamin E, 2.4 mg b-carotene, and powder or extract of fruits and berries), or a carotenoid-rich diet.

278

4. Mechanism of antioxidant action and nutrients

279 280

288 289

It is reported that antioxidants can execute protective role against free radicals by a variety of different mechanism including the catalytic systems to neutralize or divert ROS, binding or inactivation of metal ions prevents generation of ROS by Haber–Weiss reaction, suicidal and chain breaking antioxidants scavenge and destroy ROS or absorb energy, electron and quenching of ROS. In 21st century, demand for intake of antioxidant food or dietary antioxidant increasing with the hope to keep body healthy and free from diseases [78,79] and the potential beneficial effects of antioxidants in protecting against disease have been well established. It is increasingly thought that nutrition may play a vital role in helping to defend against oxidative stress and damage

t3:1 t3:2

Table 3 Effects of the health associated to the intake of Antioxidants.

t3:3

Antioxidants Vitamin C

t3:4

Vitamin E t3:5

Polyphenol t3:6 t3:7 t3:8

C E

R

R

O

286 287

C

284 285

N

282 283

induced by free radicals. Therefore, certain nutrients and dietary components with antioxidant properties are important for the protection against oxidative stress injury of the body. Food consumption is a major source of exogenous antioxidants and has been estimated that a typical diet provides more than 25,000 bioactive food constituents as nutrients and many of this may modify a multitude of processes that are related to different diseases. Generally, antioxidants are abundant in vegetables and fruits and are also found in grain cereals, peas, legumes, nuts and other food products. At this juncture, a systematic survey has also identified more than 3100 antioxidants in foods, like beverages, spices, herbs and supplements which are regularly consumed by different cultures [80]. It has been speculated that the decrease in the intake of nutritional and antioxidants rich food may

Cu, Zn, Mn, Se Other carotenoids (lycopene)

U

281

T

t2:22 t2:23 t2:24 t2:25

Effect of health

References

Protects against cancers, Protects from heart disease, Improvement of the health of cartilage, joints and skin, Maintaining a healthy immune system, Improvement in the antibody production, Increase in the absorption of nutrients, Increases protection against H2O2-induced DNA strand breaks Prevents coronary heart disease, Prevents the formation of blood clots Decreases incidence of breast and prostate cancers, Brain protection, Reduces long-term risk of dementia Decreases risk of Parkinson's disease Inhibit oxidation of LDL, Inhibit platelet aggregation, Improve endothelial dysfunction Lower risk of myocardial infarction, Effect anticarcinogenic Prevent neurodegenerative diseases, Protect against neurotoxic drugs, treatment of diabetes, treatment to prevent osteoporosis, Inhibit non-heme iron absorption Cofactors of antioxidant enzymes SOD-Cu/Zn, Mn-SOD and GSH-Pox Protection against oxidation of lipids, LDL, proteins and DNA. Abduction and free radical scavenging

Barry [85], Liu et al. [86], Wang et al. [87], Wintergerst et al. [88], Woo et al. [89], Thankachan et al. [90] and Riso et al. [91].

Pryor [92], Traber et al. [93], Weinstein et al. [94], Muller et al. [95], Devore et al. [96] and Miyake et al. [97]. Manach et al. [98], Russo et al. [99], Sch¨achinger et al. [100], Corder et al. [101], Yang et al. [102] Halliwell [103], Pan et al. [104], Zunino et al. [105], Atmaca et al. [106], Hurrell et al. [107]. Visioli F et al. [108] Visioli F et al. [108]

Please cite this article as: Rajendran P, et al, Antioxidants and human diseases, Clin Chim Acta (2014), http://dx.doi.org/10.1016/j.cca.2014.06.004

290 291 292 293 294 295 296 297 298 299 300 301 302

P. Rajendran et al. / Clinica Chimica Acta xxx (2014) xxx–xxx

310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325

329

Chronic autoimmune inflammation, which is commonly observed in rheumatoid arthritis (RA), disrupts the delicate balance between bone resorption and calcification causing the destruction of the bone and joints. Multiple aetiologies are suspected to contribute to the formation of RA, including defective articular cartilage structure and biosynthesis, joint trauma, joint instability, congenital and developmental abnormalities, and inflammatory conditions [109,110]. Oxidative stress induced damage to essential cell components caused by oxygen free radicals is a mechanism in the patho-biology of degenerative rheumatoid arthritis [109]. Hydroxyl radicals cause degradation of isolated proteoglycans, and hypochlorous acid (HOCl) fragments of collagen. Hydrogen peroxide, which is highly diffusible, readily inhibits cartilage proteoglycan synthesis, by interfering with ATP synthesis, in part by inhibiting the glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase in chondrocytes, aggravating the effects of proteolytic and free-radicalmediated cartilage degradation. Peroxynitrite and HOCl may facilitate cartilage damages by inactivating TIMPs. TIMP-1 inhibits stromelysins, collagenases and gelatinasesand this ability is lost after nitrus oxide (NO− 3 ) or HOCl treatment. HOCl can also activate latent forms of neutrophil collagenases and gelatinase with obvious consequences. − Hypochlorous acid, NO− 3 and O2 react with ascorbate, which is essential

330 331

U

N C O

R

R

E

C

T

E

D

326 327

5.1. Antioxidant vs arthritis

F

309

328

O

307 308

5. Antioxidants vs human diseases

R O

305 306

increase the chances of oxidative stress which may lead to cell damage, therefore intake of such natural antioxidants may give protective effect against free radical induced diseases [80,81]. In view of the importance of antioxidants, Suntres [82] reported that antioxidant liposomes will hold an important role in future research on antioxidants, and also reports that it facilitates the delivery of antioxidants to specific sites as well as achieving prophylactic and therapeutic action. In addition, Bouayed and Bohn [83] have suggested that the balance between oxidation and anti-oxidation is critical in maintaining a healthy biologic system and low doses of antioxidants may be favorable to this system, but high quantities may disrupt the balance. It is true that antioxidants are beneficial and display a useful role in the maintenance of the homeostasis in human ROS system, but so are pro-oxidants, therefore the scientific community should search deeper into the kinetics and in vivo mechanisms of antioxidants to uncover the optimal concentrations or desired functions in order to push forward against cancer, neurodegenerative and cardiovascular diseases [84] (Table 3). Various studies related to free radicals, oxidative stress and antioxidant activity of food reveals the prominent beneficial role of antioxidant and its specific role against different diseases individually. However the collective and concise data on the role of antioxidants in human diseases will be better and most appropriate one in order to know the exact role of antioxidants in all kinds of human diseases (Fig. 1). Hence, an attempt has been made in this review, to summarize the detailed role and impact of antioxidants in certain selected dreadful human diseases.

P

303 304

5

Q1

Fig. 1. Antioxidants/oxidative stress and health diseases.

Please cite this article as: Rajendran P, et al, Antioxidants and human diseases, Clin Chim Acta (2014), http://dx.doi.org/10.1016/j.cca.2014.06.004

332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350

6

U

Table 4 Clinical trials (variables observed: vitamins C, E, selenium, and β-carotene, ORAC).

N

t4:3

Study

t4:4

Hematologic malignancies Clemens et al. [157] Subjects: n = 19

CY, MEL, VP16, TBI

Clemens et al. [158]

Subjects: (TBI+) N = 16, mean age: 30.5 y, Controls: (TBI+) n = 10, mean age: 37.5 y Controls: n = 19, mean age: 33 y; Subjects: n = 10, mean age: 37 y Controls: n = 26, age range: 18–28 y; subjects: n = 16, intervention: n=6

CY, VP16, BU, TBI

Subjects: n = 56; controls: n = 44, mean age: 58.7 y

DXR, CY, CDDP, MELPH

Four arms: 1) VE 300 mg, Se 96 μg; 2) Se 96 μg; 3) VE 300 mg; 4) placebo (PO)

Controlled trial

Subjects: n = 18, age range: 51.8–69.6 y

CY, ADR, VP16, VCR, DXR, EPI CBPT, RT

MV β-Car 10,000–20,000 U, α-Toc 300–800 U, AA 2000– 5000 U, Se 856 μg (PO)

Nonrandomized controlled trial

Subjects: n = 32, age range: 32–81 y

Chemotx, RT, Surgery

VC 2850 mg, VE 2500 IU, β-Car 32,500 U, Se 387 μg (PO)

Nonrandomized, noncontrolled trial

Sample size and demographics

Cancer agents

C

O

t4:5

t4:6

Hunnisett et al.,[159]

t4:7

Lloyd et al. [160]

t4:8 t4:9 t4:10

Gynecologic cancer Sundstrom et al. [161]

t4:11 t4:12 t4:13

t4:14 t4:15 t4:16

t4:17 t4:18 t4:19 t4:20 t4:21 t4:22

Lung cancer Jaakola et al. [162]

Breast cancer Lockwood et al. [163]

Dosage and mode

Study design

Results

Strengths/weaknesses

α-Toc 9 mg (TPN)

Cohort study

Subjects (TBI+): serum α-Toc and β-Car 2 (baseline v day +12); TBI-α-Toc over time (P b .001), no significant changes in β-Car

Nonrandomized, nonblinded, controlled trial

↓ Hepatotoxicity; no significant differences in rates of relapse

20 sequential patients

Acute 2 in VC and VE (P b. .001; P b .001); no significant differences after day 0

Significant differences between two groups at baseline; nonrandomized, small cohort, no controls; intervention assessed the effects of RDA only Study groups received different TPN regimens; significant differences were not observed at each time point; P values NR; small cohort, different anticancer regimens; TBI+ also received 130 mg AA and 13.1 mg α-Toc by TPN Nonrandomized, small cohort; cancer diagnosis and agents NR

Cohort study

Prior to intervention: mean platelet and plasma ↓AA In CML controls (P b .001; P b .05), mean platelet AA↓ in CML v controls (P b .05); after intervention: ↑ in platelet and plasma AA in AL & CML (P b .025) ↑ plasma AA in CLL (P b .05)

Inquired about dietary intake/Cancer therapy NR; time of blood withdrawal, length of observation, and timing of intervention NR; methodology NR

↑ in serum Se in Se group 1 and group 2 v controls (P b .001; P b .001)

Placebo controlled trial/multiple gynecologic cancers, stages, and regimens

R

TBI+: α-Toc 825 mg, β-Car 45 mg, AA 450 mg, (PO); TBI+: AA 530 mg, α-Toc 9 mg (TPN)

R

E

High-dose chemotx, ±TBI

VC 25 mg; VE 1000 IU (TPN)

NR

AA 1 g X 4 days (mode NR)

C

T

E

D

P

Study group had increased survival rates compared to historical controls (P value NR); plasma Se, α-Toc NS, plasma AA, β-Car NR

R O

O

Whole blood β-Car, VE, and Se ↑ after 3 and 12 months (P b .01), P b .05), P b .01); no deaths, expected four; no subject had weight loss, ↓ use of pain killers, ↑ QOL

F

Variable doses administered depending on “individual” micronutrient analysis (methodology NR); multiple stages combined; one subject received no cancer therapy

Whole blood VC NR, no controls, chemotherapy agents NR; subjects also administered MV containing Co-Q10 and essential fatty acids; P values not consistently reported

Abbreviations: ORAC, Oxygen radical absorbance capacity; CY, cyclophosphamide; MELPH, melphalan; VP16, etoposide; 2, decrease; 1, increase; TBI, total body irradiation; α-Toc, alpha tocopherol; TPN, total parenteral nutrition; β-Car, beta carotene; RDA, recommended dietary allowances; y, years; BU, busulfan: PO, orally; AA, ascorbic acid; NR, not reported; VC, vitamin C; VE, vitamin E; DZ, diaziquone; TP, thiotepa; Ara-C, cytosine arabinoside; k-Cal, total caloric intake; CML, chronic myelocytic leukemia; AL, acute leukemia; CLL, chronic lymphocytic leukemia; VCR, vincristine; DXR, doxorubicin; EPI, epirubicin; CBPT, carboplatin; RT, radiation therapy; MV, multivitamin; NS, nonsignificant; CDDP, cisplatinum diammine dichloride; Se, selenium; chemotx, chemotherapy; QOL, quality of life; Co-Q10, coenzyme Q 10; y-TOC, gamma tocopherol (Modified from Elena J. Ladas et al., 2004).

P. Rajendran et al. / Clinica Chimica Acta xxx (2014) xxx–xxx

Please cite this article as: Rajendran P, et al, Antioxidants and human diseases, Clin Chim Acta (2014), http://dx.doi.org/10.1016/j.cca.2014.06.004

t4:1 t4:2

P. Rajendran et al. / Clinica Chimica Acta xxx (2014) xxx–xxx

372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389

394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414

C

370 371

E

F

O

368 369

R

366 367

R

364 365

N C O

362 363

U

360 361

R O

Reactive Oxygen Species is a hallmark of human cancer. ROS and their functions with respect to the cancer initiation and signalling in cancer cells is a prime concern of cancer research. Tobacco smoke also plays a very important role in increasing the risk for inflammation and cancer due to its high carcinogenic potential and the synergistic effects with other respirable particulate to generate ROS and catalyse redox reactions in the cells of humans, leading to oxidative stress and increased production of mediators of inflammation [117–119]. Inflammatory cells are particularly effective in generating most of the reactive oxygen species. The activation of the redox metabolism of the inflammatory cells generates a highly oxidative environment within an organ of aerobic organisms. Much of the oxygen biochemistry, through the activation of plasma membrane NADPH oxidase, of macrophages and neutrophils is directed towards the release of superoxide anion, hydrogen peroxide and hydroxyl radicals [120–122]. Inflammation acts through the formation of ROS and reactive nitro species (RNS) which cause oxidative damage to cellular components. Many pro-inflammatory mediators, especially cytokines, chemokines and prostagladins, turn on the angiogenesis switches mainly controlled by vascular endothelial growth factors [123]. Cancer-associated inflammation is also linked with immune-suppression that allows cancer cells to evade detection by the immune system. Inflammation is a critical component of tumor progression. Many cancers arise from sites of infection, chronic irritation

358 359

P

392 393

357

D

5.2. Anti oxidant vs cancer

355 356

and inflammation. It is now becoming clear that the tumor microenvironment, which is largely orchestrated by inflammatory cells, is an indispensable participant in the neoplastic process, fostering proliferation, survival and migration. In addition, tumor cells have co-opted some of the signalling molecules of the innate immune system such as selectins, chemokines and their receptors for invasion, migration and metastasis. These insights are fostering new anti-inflammatory therapeutic approaches to cancer development [124]. Pathological angiogenesis is a hallmark of cancer and various ischaemic and inflammatory diseases. Chronic inflammation is associated with angiogenesis, a process that helps cancer cells to grow. Angiogenesis is necessary for expansion of tumor mass and macrophage, platelets, fibroblasts and tumor cells are a major source of angiogenic factors (vascular endothelial growth factors, chemokines, NO, etc.) [125,126]. The inflammation in the tumor microenvironment is characterized by leukocyte infiltration, ranging in size, distribution and composition, as: tumorassociated macrophages (TAM), mast cells, dendritic cells, natural killer (NK) cells, neutrophils, eosinophils and lymphocytes. These cells produce a variety of cytotoxic mediators such as ROS and RNS, serine and cysteine proteases, membrane perforating agents, matrix metalloproteinase (MMP), tumor necrosis factor α (TNFα), interleukins (IL-1, IL-6, IL-8), interferons (IFNs) and enzymes, likecyclooxygenase-2 (COX-2), lipooxygenase-5 (LOX-5) and phospholipase A2 (PLA2). Other researchers discovered that tumor-associated macrophages are key regulators of the link between inflammation and carcinogenesis [127] and experimental studies showed that, tobacco smoke produce ROS that have been associated with activation of mitogen-activated protein kinase (MAPK) family members and activation of transcription factors such as NF-κβ and AP-1. These signaling pathways have been implicated in processes of inflammation, apoptosis, proliferation, transformation and differentiation [128]. The activation protein-1 (AP-1) contributes to basal gene expression in biological systems. ROS can activate AP-1 through several mechanisms and the effect of AP-1 activation is an increased cell proliferation due to increased expression of growth stimulatory genes such as cyclin D1 and suppression of the protein p21waf. Studies showed that AP-1 and NF-kB, inducible by tumor promoters of oxidative stimuli, show differential protein levels or activation in response to tumor promoters in JB6 cells. Researchers suggest that as long as oxidative events regulate AP-1 and NF-kβ transactivation, these oxidative events can be important molecular targets for cancer prevention [129,130]. NF-kB is expressed and participates in a wide range of biological processes, such as cell survival, differentiation, inflammation and growth [131]. NF-kB activation has been linked to a wide spectrum of extracellular stimuli and oxidants and subsequent involvement in the carcinogenic process through promotion of angiogenesis and tumor cell invasion and metastasis [132,133]. Dietary factors like chlorogenic acid have also been found to protect against environmental carcinogen induced carcinogenesis by their upregulation of phase II conjugating enzymes and suppression of ROSmediated correct, AP-1, and MAPK activation [134]. Cellular defence system is comprised of several Phase II detoxification enzymes such as glutathione-S-transferases (GSTs), NADP (H) quinoneoxidoreductase (NQO1), Glutathione peroxidases (GPx), Catalase, superoxide dismutases (SODs), epoxide hydrolase, hemeoxygenase (HO-1), UDPglucuronosyltransferases (UGTs), gammaglutamylcysteinesynthetase and many others. Expression of these proteins protects cells from oxidative damage and can prevent mutagenesis and cancer [135–138]. To counterbalance the oxidative damage from ROS, aerobic organisms have created a variety of antioxidant mechanisms to maintain their genomic stability. These mechanisms include phase II detoxification and other anti-oxidation enzymes that act in cellular defence such as catalase and SOD, GPx, GST, other constitutive and inducible antioxidants, DNA repair enzymes, and other cellular mechanisms of genomic surveillance, such as cell cycle checkpoint control systems [134]. Moreover, several growth factors such as serum, insulin like growth factor I, or fibroblast growth factor 2 generates ROS in MIA PaCa-2 and PANC-1

E

391

353 354

T

390

for cartilage function, leading to low levels of ascorbate in synovial fluid. Low concentrations of hydrogen peroxide (H2O2), O.− 2 or both, accelerate bone resorption by osteoclasts, whereas nitric oxide (NO.) inhibits it and promotes chondrocyte apoptosis, thereby inhibits proteoglycan synthesis and activates latent metalloproteinases and cyclooxygenase. ROS, produced by activated phagocytes, could alter the antigenic behavior of immunoglobulin G, producing fluorescent protein aggregates that can further activate phagocytic cells. Radical-exposed IgG is able to bind rheumatoid factor and results in the generation of C3a. This reaction may be self-perpetuating within the rheumatoid joint, suggesting that free radicals play a role in the chronicity of the inflammatory reaction which is a key question regarding to which extent free radicals contribute to the consequences of inflammation, such as the cartilage and bone destruction. Reactive oxygen intermediates can also function as signaling messengers to activate transcription factors, like NF-κB Change all to NF-κBand activator protein 1(AP-1), and induce gene expression [111]. All this knowledge might serve to apply a rational selection of antioxidants for possible therapeutic purposes, of the inflammatory joint disease. Many raw foods contains natural antioxidants, including enzymes such as superoxide dismutase, glutathione peroxidase and catalase, which are usually inactivated during food processing, and nonenzymic antioxidants such as carotenoids (e.g. canthaxanthin and astaxanthin in some farmed fish), β-carotene, lutein, lycopene, tocopherols (in oils) and other compounds in plants. The latter include other carotenoids and ascorbate (vitamin C). The plasma concentrations of these are largely determined by dietary intake and it is possible that two or more antioxidants can act together synergistically [112,113]. In view of the high antioxidant content of the French diet which is rich in fruit, vegetables and red wine [114,115], it is intriguing to note the relatively low incidence of arthritis [116]. More work is required on tissue distributions and bioavailability of antioxidant molecules within joints since lipophilic antioxidant molecules, such as vitamin E or β-carotene, may not have the same access to tissues as hydrophilic antioxidants, such as vitamin C. Perhaps, the differences in their effects in disease processes may depend on the hydrophilicity of the antioxidant molecules concerned and the resulting pattern of tissue distribution in different tissue areas. One major problem is that there is no assay currently available to measure oxidant activity within joints and possibly the activity could occur from an alternative mechanism.

351 352

7

Please cite this article as: Rajendran P, et al, Antioxidants and human diseases, Clin Chim Acta (2014), http://dx.doi.org/10.1016/j.cca.2014.06.004

415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480

504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546

599

Glucose overload may damage the cells through oxidative stress. This is currently the basis of the “unifying hypothesis” that hyperglycemia-induced oxidative stress may account for the pathogenesis of all diabetic complications [165,166]. Increasing evidence in both experimental and clinical studies suggests that oxidative stress plays a major role in the pathogenesis of both types of diabetes mellitus. Free radicals are formed disproportionately in diabetes by glucose oxidation, non-enzymaticglycation of proteins, and the subsequent oxidative degradation of glycatedproteins [167]. This increased superoxide production causes the activation of 5 major pathways involved in the pathogenesis of complications: polyol pathway flux, increased

600 601

O

F

5.3. Antioxidant vs diabetes

R O

502 503

P

500 501

547 548

D

498 499

cells. It has been suggested that, as a nutrition factor, POX may modulate apoptosis signals induced by p53 or other anti-cancer agents and enhance apoptosis in stress situations [148]. Components of the cell signaling network, especially those which converge on the ubiquitous eukaryotic redox-sensitive transcription factor nuclear factor-kappa B (NF-κB), have been implicated in pathogenesis of many inflammation-associated disorders. Under normal physiologic conditions, NF-κB is sequestered in the cytoplasm by binding to the inhibitory protein called IκBa. Phosphorylation and subsequent ubiquitination results in degradation of IκBa by proteasomes. Phosphorylation of IκBa is mainly mediated by the IκB kinase (IKK) complex. Phosphorylation of specific serine residues of p65 subunit of NF-κB has been considered to facilitate the translocation of NF-κB to nucleus and interaction with the coactivator CBP/p300. Induction of κphase-2 detoxifying or antioxidant genes represents an important cellular defense in response to oxidative and electrophilic insults. Nuclear transcription factor erythroid 2p45 (NF-E2)-related factor 2 (Nrf2) plays a crucial role in regulating phase-2 detoxifying/antioxidant gene induction [149]. Like NF-κB, Nrf2 is present in the cytoplasm as an inactive complex with the inhibitory protein subunit, in this case Keap1. Dissociation of Nrf2 from Keap1 is prerequisite for nuclear translocation and subsequent transactivational activity. Once translocated into nucleus, Nrf2 interacts with a small Maf protein, forming a heterodimer that binds to antioxidant responsive elements (ARE) or electrophile responsive elements (EpRE) present in the promoter/enhancer regions of genes encoding many antioxidant and detoxifying enzymes. Keap1 contains several cysteine residues that function as sensors of redox changes. Oxidation or covalent modification of these critical cysteine thiols diminishes the affinity of Nrf2 for Keap1, releasing Nrf2 for nuclear translocation. Dissociation of the Nrf2-Keap1 complex is also assumed to be stimulated through the phosphorylation of Nrf2 by distinct upstream kinases such as MAPKs, PKC, PI3K, etc. As in the case of NF-κB, phosphorylation of Nrf2 is also considered to facilitate the interaction of this redox-sensitive transcription factor with CBP/p300.NRF2 is a known regulator of the antioxidant response [150,151]. NRF2regulated phase II enzymes protect against the development of cancer by catalyzing reactions that convert highly reactive, carcinogenic chemicals to less reactive products [152,153]. Singh et al. (2012) [154] recently demonstrated that vitamin C and BHA provide protection against E2-mediated oxidative DNA damage but the mechanism is not well understood. Many antioxidants derived from dietary and medicinal plants have been found to modulate Nrf2-Keap1 signaling, thereby potentiating cellular antioxidant capacity or facilitating detoxification of carcinogens and other toxicants [155,156]. Table 4 describes 7 observational studies reporting changes in serum, plasma, or whole blood levels of vitamins Cand E, selenium,β-carotene, and total radical antioxidant parameter (TRAP), which represents total body antioxidant status, in patients undergoing anticancer therapy. Information is grouped by malignancy and includes subject demographics, cancer data, cancer treatment, end points evaluated, results, and comments. No specific treatment was consistently associated with changes in the individual antioxidants (Table 4) [164].

T

496 497

C

494 495

E

492 493

R

490 491

R

488 489

O

487

C

485 486

N

483 484

cells, which are human pancreatic adenocarcinoma cells that promotes cell survival. Inhibiting ROS generation with the antioxidants or NADPH oxidase (Nox4) antisense, or MnSODover expression would stimulate apoptosis in PaCa-2 and PANC-1 cells. This mechanism might play an important role in pancreatic cancer resistance for treatment and thus represent a novel therapeutic target. In gastrointestinal and colon cancer, oxidative pentose pathway (OPP) and the glutathione (GSH) antioxidant defense system play an important role in the regulation of cell growth and apoptosis. The OPP modulate intracellular redox status and provides NADPH for the synthesis of GSH, which is responsible for the inactivation of intracellular ROS that induce apoptosis and cell injury. Depletion of GSH increases the sensitivity of cells to ROS. Therapeutic inhibition of the OPP and/or the GSH defense system might increase the sensitivity of gastric and colon cancer cells to anti-cancer therapy [139]. Recent studies have shown that the enzymatic product of thymidine phosphorylase (TP) generated ROS within cancer cells that help in maintaining the growth of colon cancer cells thus may provide improved therapeutic results as well as a preventative effect on carcinogenesis of the colorectum [125]. Emerging evidence also suggests that supranutritional doses of selenite could induce typical apoptosis in colorectal cancer cells in vitro and in xenografttumors by inducing ROS and it indicates that antioxidants can activate the apoptotic machinery through redox-dependent activation and could be useful in cancer therapy [140]. The MAPK signalling cascade is activated by a wide variety of receptors involved in growth and differentiation including receptor tyrosine kinases (RTKs), integrins and ion channels. This pathway has been reported to be activated in over 50% of acute myelogenous leukemia, acute lymphocytic leukemia and are frequently activated in breast, prostate cancers and other cancer types [141,142]. Under optimal growth conditions, transiently elevated ROS levels confer a growth advantage to tumor cells. However, exposure of these cells to anticancer agents induces a prolonged increase in ROS levels resulting in potentiating of apoptosis. Thus ROS modulates the ability of stress kinases to stimulate cell growth or cell death and it depends on signal intensity and signal duration [143]. This idea has been nurtured to explain the complex nature of ERK signaling in cell cycle regulation and in stress activated protein kinase signalling (SAPK). Transient, reduced activity of SAPK promotes cell proliferation, whereas persistent, increased activity results in cell killing [144]. Conversely, in drug-resistant tumors, phase II enzymes or other antioxidant defences are often up regulated, shielding cells from apoptosis. ASK1, an upstream regulator of SAPKs, is inhibited in normal cells through its association with thioredoxin, which is an antioxidant capable of metabolizing ROS. Increased levels of ROS lead to the dissociation of this complex and thereby facilitate the activation of ASK1 and subsequently SAPKs [145]. A similar redox control has been identified for JNK where increased level of ROS triggers the detachment of JNK associated glutathione-S-transferase-π (GSTp) and thereby facilitating JNK activation. Moreover, ROS-dependent activation of JNK may also lead to knock down of a JNK phosphatase via an unknown mechanism linking ROS and stress induced kinases an important player in cancer cell growth [146]. Growth inhibition of pancreatic cells is dependent on the efficiency of scavenging ROS as well as effective inhibition of ERK1/2 signaling pathway activation as demonstrated by using the free radical scavenger N-Acetyl Cysteine (NAC) or the MEK/ERK1/ 2 inhibitor (PD98059). Moreover, ERK1/2 induction is dependent on ROS production as demonstrated by a complete removal of ERK1/2 phosphorylation by NAC. Thus ROS act as pro-survival, anti-apoptotic factor in pancreatic cancer cells [52]. Moreover, treatment of SW620 (colon cancer cells) with berberine, which is a major constituents of Coptidisthizoma, resulted in activation of apoptosis by phosphorylation of JNK and p38 MAPK, as well as generation of the ROS [147]. Furthermore, proline oxidase (POX), that often considered as a ‘housekeeping enzyme’, induce apoptosis through both intrinsic and extrinsic pathways and is involved in nuclear factor of activated T cells (NFAT) signaling and regulation of the MEK/ERK pathway in colon cancer

U

481 482

P. Rajendran et al. / Clinica Chimica Acta xxx (2014) xxx–xxx

E

8

Please cite this article as: Rajendran P, et al, Antioxidants and human diseases, Clin Chim Acta (2014), http://dx.doi.org/10.1016/j.cca.2014.06.004

549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598

602 603 604 605 606 607 608 609 610

9

T

E

D

P

R O

O

F

P. Rajendran et al. / Clinica Chimica Acta xxx (2014) xxx–xxx

618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640

E

R

616 617

R

614 615

formation of AGEs (advanced glycation end products), increased expression of the receptor for AGEs and its activating ligands, activation of protein kinase C isoforms, and overactivity of the hexosamine pathway. It also directly inactivates 2 critical anti-atherosclerotic enzymes, endothelial nitric oxide synthase and prostacyclin synthase. Through these pathways, increased intracellular reactive oxygen species cause defective angiogenesis in response to ischemia, activate a number of pro-inflammatory pathways, and cause long-lasting epigenetic changes that drive persistent expression of proinflammatory genes after glycemia is normalized (“hyperglycemic memory”). Atherosclerosis and cardiomyopathy in diabetes are caused in part by pathway-selective insulin resistance, which increases mitochondrial ROS production from free fatty acids and by inactivation of anti-atherosclerosis enzymes by ROS. Over expression of antioxidant enzymes in diabetic patients prevents diabetic retinopathy, nephropathy, and cardiomyopathy [168]. Antioxidants have proven to play a minimal if any role in the treatment of diabetic complications in humans [169]. Decreases in expression, and in some instances the activity of antioxidant enzymes, has been previously reported in diabetic microvascular disease [170]. Indeed, the over expression of Cu+Zn2+ superoxide dismutase (SOD) protects against end organ damage in models of type II diabetic nephropathy [171]. Other studies in mice with genetic deletions of various antioxidant enzymes have also provided insight into the specific relative contributions of Mn2+SOD [172] to the development of diabetes complications. Mn2+SOD mimetics such as MnTBAP have also shown efficacy in preventing ROS-induced injury in vitro, although the utility in vivo of such drugs may be limited [173,174]. Further strengthening a potential role for the antioxidant Mn2+SOD, specific polymorphisms of the Mn2+SOD gene are associated with the development of diabetic complications [175]. Interestingly, GPx-1-deficient mice have no increased

N C O

612 613

U

611

C

Fig. 2. Shows the involvement of cellular antioxidant enzymes in cardiovascular diseases.

risk for microvascular disease, in particular diabetic complications [176], most likely because of redundancy with respect to GPx isoforms. Over-expression of catalase in experimental models of type 2 diabetic appears to be protective the diabetic complications [177]. In contrast to Mn2+SOD, however, studies in humans have indicated no relationship between catalase gene polymorphisms that interfere with its cellular expression and the incidence of type 2 diabetic patients [178]. Kunisaki et al. [179] investigated the effects of treatments with classical antioxidants such as vitamin E,vitamin C and lipoic acid. Specifically, vitamin E normalizes retinal blood flow and protein kinase C (PKC) activity in the vascular tissue of diabetic rats. One short-term experimental study showed that high doses of vitamin C can improve some aspects of endothelial dysfunction in diabetes. Another experimental study has demonstrated that intra-peritoneal administration of αlipoic acid to streptozotocin (STZ) diabetic Wistar rats normalises TBARS levels in plasma, and the retina, liver, and pancreas [180]. Furthermore, it has been reported that α-lipoic acid leads to a decrease in the severity of diabetic neuropathy by maintaining GSH levels and/or by its direct antioxidant properties [181]. However, lipoic acid administration improved endothelial function in subjects with metabolic syndrome. In another study to determine the effects of vitamin E on oxidative stress and cell membrane fluidity in the brains of diabetesinduced experimental rats, Hong et al. [182] reported that vitamin E was found to be effective for strengthening the antioxidant defense system. They noted a reduction of the accumulation of ROS such as superoxide radicals, a decrease in the generation of oxidative damaging substances such as the carbonyl value, a significantly improved lipid composition, and maintenance of membrane fluidity in the brains of the rats. Coleman suggested that triple antioxidant therapy (vitamin E, lipoic acid and vitamin C) in diabetic volunteers attenuates the

Please cite this article as: Rajendran P, et al, Antioxidants and human diseases, Clin Chim Acta (2014), http://dx.doi.org/10.1016/j.cca.2014.06.004

641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670

696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720

726 727 728 729 730 731 732 733 734

C

694 695

E

692 693

R

690 691

R

688 689

O

686 687

C

684 685

N

682 683

U

680 681

F

The mechanisms of oxidative stress and antioxidants are in patients with atherosclerosis have been evaluated in extensive animal experiments and clinical research. There is a relationship between atherosclerotic risk factors (ARF) and increased vascular production of ROS; the most important ARFs are heredity, age, hypertension, dyslipidemia, diabetes and smoking [192]. It has been reported that in in-vitro studies an increased vascular production of ROS, particularly superoxide causes deleterious effects in cell death [193,194]. ROS and reactive nitrogen species (RNS) are closely linked to the disease process. Incomplete scavenging of ROS and RNS influences the mitochondrial lipid cardiolipin, stimulates the release of mitochondrial cytochrome c and finally

678 679

O

724 725

677

R O

5.4. Antioxidant vs arthrosclerosis

675 676

P

723

673 674

activates the intrinsic death pathway [195]. Local generation of RNS may contribute to vascular tissue injury. Therefore, ROS and RNS participate as signalling molecules that regulate diverse pathophysiological signalling pathways. High levels of ROS are potent inducers of the intrinsic apoptotic pathway and tissue injury in pathophysiological conditions as an integral part of atherosclerotic plaque stabilization. The most important sources of ROS production associated with CVD pathology are the mitochondrial respiratory chain, nicotinamide adenine dinucleotide phosphate oxidases (NADP oxidase). The pathogenesis of atherosclerosis is further related to inflammation, immune response and the proliferative process. Endothelial denuding injury leads to platelet aggregation and releases platelet-derived growth factor, which triggers the proliferation of smooth muscle cells forming the nidus of the atherosclerotic plaque in the arterial intima, implicating inflammatory changes in the development of the disease [196,197]. Supplementing treatment with antioxidants might be helpful to patients. Vitamin E reduces the consequences of lipid peroxide content in mouse peritoneal macrophages (MPMs) [198]. E-selectin has been proposed as an important factor in the development of the inflammatory process underlying atherothrombosis. The expression of E-selectin is decreased by vitamin E in the diabetic rats [199]. Vitamin C has also been investigated in studies on the prevention of atherosclerosis. The following proposals have been put forward regarding the mechanisms of vitamin C in preventing atherosclerosis: First, vitamin C has been shown to prevent apoptosis caused by cytokines in cultured endothelial cells [200]. It also decreases the release of micro-particles derived from endothelial cells and suppresses pro-apoptotic activity in congestive heart failure patients in vivo [201]. Second, vitamin C stimulates all types of collagen synthesis by specific hydroxylase enzymes [202]. Endothelial cell proliferation is, in part, associated with the synthesis of type IV collagen [203]. Thus, the lack of vitamin C prevents the generation of type IV collagen in cultured endothelial cells [204]. Third, vitamin C protects the vascular endothelium by enhancing endothelial NO synthase. Endothelial NO synthase activity is inhibited by ROS that oxidize and deplete the co-factor tetrahydrobiopterin [205]. Therefore, vitamin C prevents the loss of NO synthase activity by maintaining tetrahydrobiopterin [206]. Glutathione monoethyl ester, but not ascorbic acid, exerted protective effects against ischemia–reperfusion injury. Interestingly, the protective effects of glutathione monoethyl ester are enhanced by co-administration with vitamin C in rat hearts subjected to ischemia and reperfusion [207]. The oxidized LDL leads to increased platelet-endothelial cell adhesion, which can be prevented by superoxide dismutase (SOD) and catalase [208]. In fact, the vascular extracellular expression of SOD is stimulated by NO [209]. Narang et al. [210] reported that dietary palmolein oil, which is rich in monounsaturated fatty acid and antioxidant vitamins, “protected rat heart from oxidative stress associated with ischemiareperfusion injury”. Das et al. [211] found “a role for c-Src [a family of proto-oncogenic tyrosine kinases] in post ischemic cardiac injury and dysfunction and demonstrate direct cardio protective effects of [the tocotrienol-rich fraction of palm oil (TRF)]. The cardio protectiveproperties of TRF appear to be due to inhibition of c-Src activation and proteasome stabilization”. Carlson et al. [212] using a rat model, reported: “Antioxidant vitamin therapy [vitamin C, vitamin E, vitamin A and zinc] abrogated myocardial inflammatory cytokine signaling and attenuated sepsis-related contractile dysfunction, suggesting that antioxidant vitamin therapy may be a potential approach to treat injury and disease states characterized by myocardial dysfunction”. Hypertension is directly regulated by the kidneys and cardiovascular system and adversely affects these organs. Tian et al. [213] found that in salt sensitive rat model of hypertension, vitamin C and vitamin E treatments “decreased renal inflammatory cytokines and chemokines, renal immune cells, NF-κB, and arterial pressure and improved renal function and damage”. As noted above, chronic zidovudine administration promotes cardiovascular damage, and vitamin C has been found to combat this effect in rats [214]. Diabetes mellitus may initiate increased myocardial vulnerability to ischemia–

T

721 722

experimental oxidative stress of met-hemoglobin formation in vitro and reduces haemoglobin glycation in vivo [183]. Panjwani et al. [184] suggested that vitamin C administered alone or in combination with vitamin E reduced the fall in ulnar nerve conduction velocity. Sivan and Reece [185] investigated whether dietary supplementation with vitamin E would reduce the incidence of diabetic embryopathy in an in vivo rat model. Fifty pregnant rats were designated as the control group and received a normal diet while the diabetic group received vitamin E supplements (400 mg/day). This experimental study found that supplementation with vitamin E reduces the incidence of neural tube defects by more than 75% [186]. These findings suggest that vitamin E reduces this oxidative load, confers a protective effect against diabetic embryopathy, and thus may potentially serve as a dietary prophylaxis in the future. These results are in line with the reports of Chang et al. [186] which showed that diabetic embryopathy of rat or mouse embryos is prevented by vitamin C, vitamin E, SOD, N-acetyl-cysteine, or glutathione ethyl ester. Another study by Otero et al. [187] showed the effects of supplementation with vitamin E to diabetic mice. The beneficial effect of vitamin E observed in this model was shown to retard coronary atherosclerosis accelerated by DM, and was demonstrated to be due to a reduction in oxidative stress, and not secondary to a decrease in plasma glucose or cholesterol since their respective plasma concentrations remained unchanged in the diabetic mice supplemented with vitamin E [188]. Furthermore, it has been recently reported that macrophages from diabetic mice are under excess oxidative stress, and that the antioxidant vitamin E can attenuate macrophage oxidative stress which exists in DM and leads to accelerated atherosclerosis development. In a placebocontrolled, randomized trial of diabetic patients, Reaven investigated the effect of 1600 IU of RRR-α-tocopherol supplementation daily for 10 weeks on hyperglycemia-induced LDL modifications. The result of the study pointed to a reduction of approximately 60% of plasma LDL oxidation in diabetic patients, which was statistically significant when compared to healthy controls [189]. These results are supported and confirmed by other similar clinical data. Salonen et al. [190] found that doses equal to or higher than 450 IU are sufficient to significantly ameliorate the susceptibility of LDL to oxidation, indicating that relatively high doses of RRR-α-tocopherol for supplementation are needed. Furthermore, Bellomo and others have shown that the effects of RRRα-tocopherolsupplementation on LDL oxidation are accompanied by a concomitant reduction in autoantibody levels againsthyperglycemiainduced LDL modifications. Moreover, clinical studies have shown an inhibitory effect of RRR-α-tocopherol supplementation on thehyperglycemia-induced LDL modifications inT1DM and T2DM diabetic patients. Based on nutrition recommendations and interventions for diabetes, there is no clear evidence of benefits that can be derived from vitamin or mineral supplementation in people with diabetes. Routine supplementation with antioxidants, such as vitamins E and C and carotene, is not advised because of lack of evidence of efficacy, and concern related to long term safety, and therefore cannot be recommended [191]. The majority of studies included in this review support a possible role of antioxidant supplementation in reducing the risk of diabetic complications

D

671 672

P. Rajendran et al. / Clinica Chimica Acta xxx (2014) xxx–xxx

E

10

Please cite this article as: Rajendran P, et al, Antioxidants and human diseases, Clin Chim Acta (2014), http://dx.doi.org/10.1016/j.cca.2014.06.004

735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800

P. Rajendran et al. / Clinica Chimica Acta xxx (2014) xxx–xxx

825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864

C

823 824

E

821 822

R

819 820

R

817 818

N C O

815 816

U

6. Conclusion

910

In summary the reactive oxygen species and oxidative stress play an important role in the aetiology and progression of major human degenerative diseases has triggered enormous and worldwide interest in endogenous and exogenous antioxidants. There is now abundant evidence that substances in fruit and vegetables are potent preventives of various diseases, especially arthritis, cancer, heart disease, diabetes and neurodegenerative diseases. With these recent developments in scientific knowledge which firmly establish the links between food factors and the prevention of disease. The present review reveals a vast number of relevant studies related to oxidative stress and selected human diseases. The research field actually presents a variety of approaches that have been produced suggestive evidence that antioxidants might have an impact against many diseases. However, more systematic research in this line is imperative in order to reveal the relationship and involvement of antioxidants with other diseases. The large diversity in the molecular mechanisms and the methodology of antioxidants discussed here are factors probably responsible for the consistent findings of their prevention of diseases. This shows that it will be very

911

F

813 814

O

Neurodegenerative disorders are a heterogeneous group of diseases of the nervous system, including the brain, spinal cord, and peripheral nerves that have much different aetiology. ROS are particularly active in the brain and neuronal tissue as the excitatory amino acids and neurotransmitters, whose metabolism is factory of ROS, which are unique to the brain and serve as sources of oxidative stress. ROS attack glial cells and neurons, which are post-mitotic cells and therefore, they are particularly sensitive to free radicals, leading to neuronal damage [228]. It has been reported that deleterious effects of ROS on human cells may end in oxidative injury leading to programmed cell death i.e. apoptosis [229]. A multitude of genes involved in redox status, antiinflammation and detoxification are transcribed by Nrf2–ARE pathway activation. These genes are known to be involved in cytoprotection from various oxidative insult and cellular injuries in numerous different tissues and organs including brain. Intracellular peroxidases are cleared by a group of enzymes that are transcribed by Nrf2–ARE pathway. The peroxisomal CAT catalyses the conversion of H2O2 to water and molecular oxygen. However, the specific activity of CAT is much lower in brain than peripheral tissue [230]. Glutathione-s-peroxidase (Gpx) is another enzyme that metabolizes H2O2 and depends on reduced glutathione (GSH). The oxidized GSH (GS-SG) is recycled to GSH by glutathione reductase (GR). Glutathione (GSH), a tripeptide γ-glutamyl-cysteinylglycine, is the most abundant low molecular weight thiol expressed ubiquitously. It is widely recognized as an endogenous non-enzymatic antioxidant and an oxyradical scavenger, and is thereby critical to maintaining a reducing environment in the cell and protect against oxidative damage by ROS [231–235]. GSH has been implicated in a wide range of metabolic processes, including cell division, DNA repair, regulation of enzyme activity, and activation of transcription factors, modulation of anion and cation homeostasis, and protection against oxidative damage [236]. GSTs are key detoxification enzymes that catalyze the conjugation of various electrophiles, reactive alkenals, and numerous other xenobiotic to GSH. These GSH-S-conjugates are removed from cells by the multidrug resistant protein-1 (MRP-1), an ATP binding cassette (ABC) family protein [237,238]. MRP-1 is an integral plasma membrane protein that exports glutathione-S conjugates out of the cell in an ATP-dependent manner [239,240]. Studies have shown reduced GST activity in brain and ventricular fluids in AD [241]. Increased expression of GST leads to increased resistance towards oxidative stress in neuroblastoma cells and provides protection against HNE-mediated toxicity in neuronal cell culture [241]. Growing evidence demonstrates that the AD brain is under tremendous oxidative stress. A significantly increased HO-1 expression was reported in post-mortem AD temporal cortex and hippocampus compared to aged-matched control [242]. Additionally, an increased Nqo1 activity and expression was found in astrocytes and neurons of AD brain [231] and Nrf2 was predominantly localized in cytoplasm in AD hippocampal neurons [243]. Furthermore, there is increased protein oxidation [244,245] and lipid peroxidation [246–248] in AD brain when compared to aged matched controls. Recent studies in aged APP/PS1 AD mouse models showed reduced Nrf2, Nqo1, GCL catalytic subunit (GCLC) and GCL modifier subunit (GCLM) mRNA and Nrf2 protein levels [249]. Additionally, in a triple transgenic AD mouse, the GSH/GSSG ratio was reported to be reduced [250].

R O

811 812

807

865 866

P

5.5. Anti oxidant vs neurodegerative diseases

805 806

The hallmark of PD is a severe reduction of dopamine in all components of the basal ganglia. Dopamine and its metabolites are depleted in the caudate nucleus, putamen, globuspallidus, nucleus accumbens, the ventral tegmental area, and the substantianigra pars compacta and reticulata. Moderate losses of dopamine are found in the lateral hypothalamus, medial olfactory region and amygdaloid nucleus [251]. In early parkinsonism, there appears to be a compensatory increase in dopamine receptors to accommodate the initial loss of dopamine neurons [252]. As the disease progresses, the number of dopamine receptors are decreases, apparently due to the concomitant degeneration of dopamine target sites on striatal neurons. In the remaining neurons in patients with PD, dopamine turnover seems greatly increased, judging from the concentrations of homovannilic acid [HVA] in the nerve terminals in the striatum and the cell bodies and dendrites in the substantia nigra [253] and the ROS production may very well increase in consequence. This hypothesis is strengthened by a study showing that the concentrations of GSH decrease when dopamine turnover increase after reserpine treatment in rats, indicating increased activity of the peroxide scavenging enzyme GSH-Px [254]. If the increase in ROS production due to increased dopamine turnover is not buffered by the scavenging enzymes (SOD, catalase, and GSH-Px), the compensatory hyperactivity of the dopaminergic neurons may become selfdestructive. Chronic administration of L-DOPA would then only exacerbate the production of destructive ROS [255]. The administration of L-DOPA itself has been postulated to enhance the accumulation of ROS [256]. Another index of oxidative stress in PD might be the evidence of a robust increase of NF-κB in the nuclei of dopaminergic neurons in the substantia nigra of PD patients [257]. This clinical finding is consistent with in vitro data showing that oxidative stress induced by C2ceramide treatment causes nuclear translocation of NF-κB in cultured mesencephalic neurons [258]. More recently, it has been shown that the neurotoxin 6-OHDA activates NF-κB in PC12 cells by enhancing intracellular ROS levels [259]. Interestingly, in this experimental model, NF-κB seems to sustain cell survival by stimulating the expression of the anti-apoptotic proteins Bcl-2 and Bfl-1144. Moreover, as already mentioned, the potent green tea polyphenol antioxidant EGCG exerts a neuroprotective effect in a MPTP mouse model of PD [260]. However, there are still few and controversial epidemiological data on this important point, which might be partly due to the intrinsic difficulties in performing epidemiological surveys regarding the dietary habits of large populations. Nevertheless, it is desirable that future studies aimed at investigating the relationship between dietary antioxidant intake and the relative risk for neurodegenerative disorders such as AD, PD, and ALS will throw more light on this very important aspect of public health.

D

810

803 804

T

808 809

reperfusion injury and pro/antioxidant imbalance. Resistance to ischemia-induced ventricular arrhythmias and levels of endogenous antioxidants (α-tocopherol) has been found to be increased in diabetic rat myocardium [215]. The currently available evidence needs to be confirmed in clinical trials. Consumption of fruit and vegetable diets rich in antioxidant nutrients can be recommended for all individuals, and there is some indication that such a diet can be beneficial in the effects of cardiovascular events. Fig. 2 shows the involvement of cellular antioxidant enzymes in cardiovascular diseases.

E

801 802

11

Please cite this article as: Rajendran P, et al, Antioxidants and human diseases, Clin Chim Acta (2014), http://dx.doi.org/10.1016/j.cca.2014.06.004

867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909

912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928

7. Uncited references

933 934 935 936 937 938 939 940 941

[24]

[25]

[26]

[27]

[28] [29]

[30]

[32]

[33] [34]

[35]

D

[36]

E

[37] [38] [39]

O

R

R

E

C

T

[1] Dinstel RR, Cascio J, Koukel S. The antioxidant level of Alaska's wild berries: high, higher and highest. Int J Circumpolar Health 2013;72:796–802. [2] Pham-Huy LA, He H, Pham-Huy C. Free radicals, antioxidants in disease and health. Int J Biomed Sci 2008;4:89–96. [3] Valko M, Leibfritz D, Moncol J, Cronin MTD, Mazur M, Telser J. Free radicals and antioxidants in normal physiological functions and human disease. Int J Biochem Cell Biol 2007;39:44–84. [4] Singh PP, Chandra A, Mahdi F, Roy A, Sharma P. Reconvene and reconnect the antioxidant hypothesis in human health and disease. Indian J Clin Biochem 2010;25:225–43. [5] Sen S, Chakraborty R, Sridhar C, Reddy YSR, De B. Free radicals, antioxidants, diseases and phytomedicines: current status and future prospect. Int J Pharm Sci Rev Res 2010;3:91–100. [6] Lykkesfeldt J, Svendsen O. Oxidants and antioxidants in disease: oxidative stress in farm animals. Vet J 2007;173:502–11. [7] Miller JK, Brzezinska-Slebodzinska E, Madsen FC. Oxidative stress, antioxidants, and animal function. J Dairy Sci 1993;76:2812–23. [8] Sugino N. Roles of reactive oxygen species in the corpus luteum. Anim Sci J 2006;77:556–65. [9] Reilly PM, Schiller HJ, Bulkley GB. Pharmacologic approach to tissue injury mediated by free radicals and other reactive oxygen metabolites. Am J Surg 1991;161:488–503. [10] Alberti A, Bolognini L, Macciantelli D, Caratelli M. The radical cation of N, N-Diethylpara-phenylendiamine: a possible indicator of oxidative stress in biological samples. Res Chem Intermed 2000;26:253–67. [11] Di Trana A, Celi P, Claps S, Fedele V, Rubino R. The effect of hot season and nutrition on the oxidative status and metabolic profile in dairy goats during mid lactation. Anim Sci 2006;82:717–22. [12] Rizzo A, Mutinati M, Spedicato M, et al. First demonstration of an increased serum level of reactive oxygen species during the peripartal period in the ewes. Immunopharmacol Immunotoxicol 2008;30:741–6. [13] Bernabucci U, Ronchi B, Lacetera N, Nardone A. Influence of body condition score on relationships between metabolic status and oxidative stress in periparturient dairy cows. J Dairy Sci 2005;88:2017–26. [14] Merlo M, Celi P, Barbato O, Gabai G. Relationships between oxidative status and pregnancy outcome in dairy cows. Anim Prod Aust 2008;27:84. [15] Celi P, Sullivan M, Evans D. The stability of the reactive oxygen metabolites (d-ROMs) and biological antioxidant potential (BAP) tests on stored horse blood. Vet J 2010;183:217–8. [16] Fialová L, Malbohan I, Kalousová M, et al. Oxidative stress and inflammation in pregnancy. Scand J Clin Lab Invest 2006;66:121–7. [17] Noyan T, Güler A, Sekeroglu MR, Kamaci M. Serum advanced oxidation protein products, myeloperoxidase and ascorbic acid in pre-eclampsia and eclampsia. Aust N Z J Obstet Gynaecol 2006;46:486–91. [18] Witko-Sarsat V, Friedlander M, Nguyen Khoa T, et al. Advanced oxidation protein products as novel mediators of inflammation and monocyte activation in chronic renal failure. J Immunol 1998;161:2524–32. [19] Kalousová M, Skrha J, Zima T. Advanced glycation end-products and advanced oxidation protein products in patients with diabetes mellitus. Physiol Res 2002;51:597–604. [20] Shi XY, Hou FF, Niu HX, et al. Advanced oxidation protein products promote inflammation in diabetic kidney through activation of renal nicotinamide adenine dinucleotide phosphate oxidase. Endocrinology 2008;149:1829–39. [21] Kaneda H, Taguchi J, Ogasawara K, Aizawa T, Ohno M. Increased level of advanced oxidation protein products in patients with coronary artery disease. Atherosclerosis 2002;162:221–5. [22] Atabek ME, Keskin M, Yazici C, et al. Protein oxidation in obesity and insulin resistance. Eur J Pediatr 2006;165:753–6. [23] Celi P, Raadsma HW. The effects of Yerba Mate (Ilex paraguarensis) supplementation on the productive performance of lambs. Ruminant physiology digestion,

C

946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962 963 964 965 966 967 968 969 970 971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003 1004

[31]

References

N

945

[216,217,218,219,220,221,222,223,224,225,226,227]

U

944

F

943 Q3

931 932

metabolism, and effects of nutrition on reproduction and welfare. , Proceeding of the XIth International Symposium in Ruminant PhysiologyFrance: ClermontFerrand; 2009. p. 804–5. Celi P, Raadsma HW. Effects of Yerba Mate (Ilex paraguariensis) supplementation on the productive performance of dairy cows during mid-lactation. Anim Prod Sci 2010;50:339–44. Kalousová M, Zima T, Tesar V, Dusilová-Sulková S, Skrha J. Advanced glycoxidation end products in chronic diseasesclinical chemistry and genetic background. Mutat Res 2005;579:37–46. Witko-Sarsat V, Nguyen-Khoa T, Jungers P, Drüeke TB, Descamps-Latscha B. Advanced oxidation protein products as a novel molecular basis of oxidative stress in uraemia. Nephrol Dial Transplant 1999;14(1):76–8. Merlo M, Barbato O, Stefani A, Celi P, Da Dalt L, Gabai G. Embryonic mortality and plasma advanced oxidation protein products (AOPP) increase in dairy cows. Current Topics in Dairy Production, Proceedings of the Dairy Research Foundation Symposium; 2008. p. 49–53 [Camden, Australia]. Georgieva NV. Oxidative stress as a factor of disrupted ecological oxidative balance in biological systems—a review. Bulg J Vet Med 2005;8:1–11. Janero DR. Malondialdehyde and thiobarbituric acid-reactivity as diagnostic indices of lipid peroxidation and peroxidative tissue injury. Free Radic Biol Med 1990;9:515–40. Halliwell B, Chirico S. Lipid peroxidation: its mechanism, measurement, and significance. Am J Clin Nutr 1993;57:715S–24S. Castillo C, Hernández J, Valverde I, et al. Plasma malonaldehyde (MDA) and total antioxidant status (TAS) during lactation in dairy cows. Res Vet Sci 2006;80:133–9. Bouwstra RJ, Goselink RM, Dobbelaar P, Nielen M, Newbold JR, van Werven T. The relationship between oxidative damage and vitamin E concentration in blood, milk, and liver tissue from vitamin E supplemented and nonsupplemented periparturient heifers. J Dairy Sci 2008;91:977–87. Griffiths HR, Moller L, Bartosz G, et al. Biomarkers. Mol Aspects Med 2002;23:101–208. Lykkesfeldt J. Determination of malondialdehyde as dithiobarbituric acid adduct in biological samples by HPLC with fluorescence detection: comparison with ultraviolet–visible spectrophotometry. Clin Chem 2001;47:1725–7. Milne GL, Musiek ES, Morrow JD. F2-Isoprostanes as markers of oxidative stress in vivo: an overview. Biomarkers 2005;10:10–23. Mohanty JG, Bhamidipaty S, Evans MK, Rifkind JM. A fluorimetric semi-microplate format assay of protein carbonyls in blood plasma. Anal Biochem 2010;400(2):289–94. Ohkawa H, Ohishi N, Yagi K. Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem 1979;95(2):351–8. Collins CE, Quaggiotto P, Wood L, O'Loughlin EV, Henry RL, Garg ML. Elevated plasma levels of F2α isoprostane in cystic fibrosis. Lipids 1999;6(34):551–6. Halliwell B, Gutteridge JMC. Free radical in biology and medicine. 4th ed. New York, NY, USA: Oxford University Press; 2007. Benzie IFF, Strain JJ. The ferric reducing ability of plasma (FRAP) as a measure of “antioxidant power”: the FRAP assay. Anal Biochem 1996;239(1):70–6. Roe HJ, Kuether CA. Detection of ascorbic acid in whole blood and urine through the 2, 4-dinitrophenyl-hydrazine derivative of dehydro ascorbic acid. J Biol Chem 1943;147:399–407. Teissier E, Walters-Laporte E, Duhem C, Luc G, Fruchart JC, Duriez P. Rapid quantification of alpha-tocopherol in plasma and low- and high-density lipoproteins. Clin Chem 1996;42(1):430–5. Misra HP, Fridovich I. Therole of superoxide anion in the autoxidation of epinephrine and a simple assay for superoxide dismutase. J Biol Chem 1972;247(10):3170–5. Aebi M. Catalase. In: Bergmeyer HU, editor. Methods of enzymatic analysis, vol. 2. NY, USA: Verlag Chemie-Academic Press; 1974. Flohe L, Gunzler WA. Assays of glutathione peroxidise. Met Enzymol 1984;105:114–20. Hissin PJ, Hilf R. A fluorometric method for determination of oxidized and reduced glutathione in tissues. Anal Biochem 1976;74(1):214–26. Haining JL, Legan JS. Fluorometric assay for xanthine oxidase. Anal Biochem 1967;21(3):337–43. Nauseef WM. Biological roles for the NOX family NADPH oxidases. J Biol Chem 2008;283(25):16961–5. Burger D, Touyz RM. Cellular biomarkers of endothelial health: microparticles, endothelial progenitor cells, and circulating endothelial cells. J Am Soc Hypertens 2012;6(2):85–99. Touyz RM, Schiffrin E. Arterial hypertension. In: Hill J, editor. Muscle fundamental biology and mechanisms of disease, vol. 2. San Diego, Calif, USA: Elsevier; 2012. p. 1311–9. Sinha MK, Gaze DC, Tippins JR, Collinson PO, Kaski JC. Ischemia modified albumin is a sensitive marker of myocardial ischemia after percutaneous coronary intervention. Circulation 2003;107(19):2403–5. Kohen R, Nyska A. Oxidation of biological systems: oxidative stress phenomena, antioxidants, redox reactions, and methods for their quantification. Toxicol Pathol 2002;30(6):620–50. Scholz RW, Graham KS, Gumpricht E, Reddy CC. Mechanism of interaction of vitamin E and glutathione in the protection against membrane lipid peroxidation. Ann N Y Acad Sci 1989;570:514–7. Harrison R. Structure and function of xanthine oxidoreductase: where are we now? Free Radic Biol Med 2002;33(6):774–97. Yamazaki T, Kawai C, Yamauchi A, Kuribayashi F. A highly sensitive chemiluminescence assay for superoxide detection and chronic granulomatous disease diagnosis. Trop Med Health 2011;39(2):41–5. Nauseff WM. Detection of superoxide anion and hydrogen peroxide production by cellular NADPH oxidases. Biochim Biophys Acta 2014 [in press]. Moller P, Loft S. Dietary antioxidants and beneficial effect on oxidatively damaged DNA. Free Radic Biol Med 2006;41:388–415.

O

942

useful in controlling many oxidative stress mediated diseases as discussed above. In comparison with the effects of the antioxidants analysed in the previous study we can expect that many plant derived compounds which have excellent antioxidant property and were successfully investigated here and validates or corroborates the antioxidant hypothesis of many diseases [167,261–265]. Current review highlights the perspectives and possibilities of unrevealing the uncapped potentials of antioxidants in herbal plants for disease cure and warrants the need for systematic research in the field to clear the insights in this very complex matter. However, recent research work with antioxidant therapy suggests another promising area i.e., combining potential antioxidants and using them during the early onset of the disease as a prophylactic measure to prevent the disease that may eventually prove to be more effective in treating several devastating diseases.

R O

929 930

P. Rajendran et al. / Clinica Chimica Acta xxx (2014) xxx–xxx

P

12

[40] [41]

[42]

[43] [44] [45] [46] [47] [48] [49]

[50]

[51]

[52]

[53]

[54] [55]

[56] [57]

Please cite this article as: Rajendran P, et al, Antioxidants and human diseases, Clin Chim Acta (2014), http://dx.doi.org/10.1016/j.cca.2014.06.004

1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 Q4 1018 1019 1020 1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060 1061 1062 1063 1064 1065 1066 1067 Q5 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080 1081 1082 1083 1084 1085 1086 1087 Q6 1088 1089 1090

P. Rajendran et al. / Clinica Chimica Acta xxx (2014) xxx–xxx

N C O

R

R

E

C

D

P

R O

O

F

[90] Thankachan P, Walczyk T, Muthayya S, Kurpad AV, Hurrell RF. Iron absorption in young Indian women: the interaction of iron status with the influence of tea and ascorbic acid. Am J Clin Nutr 2008;87:881–6. [91] Riso P, Martini D, Moller P, et al. DNA damage and repair activity after broccoli intake in young healthy smokers. Mutagenesis 2010:1–8. [92] Pryor WL. Vitamin E and heart disease: basic science to clinical intervention trials. Free Radic Biol Med 2000;28:141–61. [93] Traber MG, Frei B, Beckman JS. Vitamin E revisited: do new data validate for chronic disease prevention? Curr Opin Lipidol 2008;19:30–8. [94] Weinstein SJ, Wright ME, Lawson KA, et al. Serum and dietary vitamin E in relation to prostate cancer risk. Cancer Epidemiol Biomarkers Prev 2007;16:1253–8. [95] Muller DPR. Vitamin E and neurological functions. Mol Nutr Food Res 2010;54:1–9. [96] Devore EE, Grodstein F, van Rooij FJ, et al. Dietary antioxidant and lonterm risk of dementia. Arch Neurol 2010;67:819–25. [97] Miyake Y, Fukushima W, Tanaka K, et al. Dietary intake of antioxidant vitamins and risk of Parkinson's disease: a case–control study in Japan. Eur J Neurol 2010. http:// dx.doi.org/10.1111/j.1468-1331.2010. 03088. [98] Manach C, Mazur A, Scalbert A. Polyphenols and prevention of cardiovascular diseases. Curr Opin Lipidol 2005;16:77–84. [99] Russo P, Tedesco I, Russo M, Russo GL, Venezia A, Cicala C. Effects of de-alcoholated red wine and its phenolic fractions on platelet aggregation. Nutr Metab Cardiovasc Dis 2001;11:25–9. [100] Schachinger V, Britten MB, Zeiher AM. Prognostic impact of coronary vasodilator dysfunction on adverse long-term outcome of coronary heart disease. Circulation 2000;101:1899–906. [101] Corder R, Mullen W, Khan NQ, et al. Oenology: red wine procyanidins and vascular health. Nature 2006;444:566. [102] Yang CS, Landau JM, Huang MT, Newmark HL. Inhibition of carcinogenesis by dietary polyphenolic compounds. Ann Rev Nutr 2001;21:381–406. [103] Halliwell B. Role of free radicals in the neurodegenerative diseases: therapeutic implications for antioxidant treatment. Drugs Aging 2001;18:685–716. [104] Pan T, Jankovic J, Le W. Potential therapeutic properties of green tea polyphenols in Parkinson's disease. Drugs Aging 2003;20:711–21. [105] Zunino SJ, Storms DH, Stephensen CB. Diets rich in polyphenols and vitamin A inhibit the development of Type I autoimmune diabetes in nonobese diabetic mice. J Nutr 2007;137:1216–21. [106] Atmaca A, Kleerekoper M, Bayraktar M, Kucuk O. Soy isoflavones in the management of postmenopausal osteoporosis. Menopause 2008;15:748–57. [107] Hurrell RF, Reddy M, Cook JD. Inhibition of non-haem iron absorption in man by polyphenolic-containing beverages. Br J Nutr 1999;81:289–95. [108] Visioli F, Grande S, Bogani P, Galli C. The role of antioxidants in the mediterranean diets: focus on cancer. Eur J Cancer Prev 2004;13:337–43. [109] de Oliveira El-Warrak A, Rouma M, Amoroso A, Boysen SR, Chorfi Y. Measurement of vitamin A, vitamin E, selenium, and L-lactate in dogs with and without osteoarthritis secondary to ruptured cranial cruciate ligament. Can Vet J 2012;53:1285–8 [La revue veterinairecanadienne]. [110] Roush JK, McLaughlin RM, Radlinsky MAG. Understanding the pathophysiology of osteoarthritis. Vet Med 2002;97:108–17. [111] Hadjigogos K. The role of free radicals in the pathogenesis of rheumatoid arthritis. Panminerva Med 2003;45:7–13. [112] Liu C, Russell RM, Wang XD. Alpha-Tocopherol and ascorbic acid decrease the production of beta-apo-carotenals and increase the formation of retinoids from beta-carotene in the lung tissues of cigarette smoke-exposed ferrets in vitro. J Nutr 2004;134:426–30. [113] Pryor WA. Vitamin E and heart disease: basic science to clinical intervention trials. Free Radic Biol Med 2000;28:141–64. [114] Ortiz J, Marin R, Noriega D, Navarro M, Arozarena I. Color, phenolics, and antioxidant activity of blackberry (RubusglaucusBenth.), blueberry (VacciniumfloribundumKunth.), and apple wines from Ecuador. J Food Sci 2013;78:C985–93. [115] Pastor-Valero M. Fruit and vegetable intake and vitamins C and E are associated with a reduced prevalence of cataract in a Spanish Mediterranean population. BMC Ophthalmol 2013;13:52. [116] Elmali N, Baysal O, Harma A, Esenkaya I, Mizrak B. Effects of resveratrol in inflammatory arthritis. Inflammation 2007;30:1–6. [117] Gardi C, Valacchi G. Cigarette smoke and ozone effect on murine inflammatory responses. In: Valacchi G, editor. Environmental Stressors in Biology and Medicine; 2012. p. 104–11. [118] Sangani RG, Ghio AJ. Lung injury after cigarette smoking is particle related. Int J Chron Obstruct Pulmon Dis 2011;6:191–8. [119] Moller P, Folkmann JK, Forchhammer L, et al. Air pollution, oxidative damage to DNA, and carcinogenesis. Cancer Lett 2008;266:84–97. [120] Lazennec G, Richmond A. Chemokines and chemokine receptors: new insights into cancer-related inflammation. Trends Mol Med 2010;16:133–44. [121] Rodriguez-Vita J, Lawrence T. The resolution of inflammation and cancer. Cytokine Growth Factor Rev 2010;21:61–5. [122] Porta C, Larghi P, Rimoldi M, et al. Cellular and molecular pathways linking inflammation and cancer. Immunobiology 2009;214:761–77. [123] Costa C, Incio J, Soares R. Angiogenesis and chronic inflammation: cause or consequence? Angiogenesis 2007;10:149–66. [124] Valavanidis A, Vlachogianni T, Fiotakis K, Loridas S. Pulmonary oxidative stress, inflammation and cancer: respirable particulate matter, fibrous dusts and ozone as major causes of lung carcinogenesis through reactive oxygen species mechanisms. Int J Environ Res Public Health 2013;10:3886–907. [125] Allavena P, Sica A, Solinas G, Porta C, Mantovani A. The inflammatory microenvironment in tumor progression: the role of tumor-associated macrophages. Crit Rev Oncol Hematol 2008;66:1–9.

E

T

[58] Moller P. The alkaline comet assay: towards validation in biomonitoring of DNA damaging exposures. Basic Clin Pharmacol Toxicol 2006;98:336–45. [59] Boyle SP, Dobson VL, Duthie SJ, Kyle JAM, Collins AR. Absorption and protective effects of flavonoid glycosides from an onion meal. Eur J Nutr 2000;39:213–23. [60] Moller P, Loft S. Oxidative DNA damage in white blood cells of humans in dietary antioxidant intervention studies. Am J Clin Nutr 2002;76:303–10. [61] Sacheck JM, Milbury PE, Cannon JG, Roubenoff R, Blumberg JB. Effect of vitamin E and eccentric exercise on selected biomarkers of oxidative stress in young and elderly men. Free Radic Biol Med 2003;34:1575–88. [62] Collins AR, Cadet J, Moller L, Poulsen HE, Vina J. Are we sure we know how to measure 8-oxo-7,8-dihydroguanine in DNA from human cells? Arch Biochem Biophys 2004;423:57–65. [63] Arendt BM, Ellinger S, Kekic K, et al. Single and repeated moderate consumption of native or dealcoholized red wine show different effects on antioxidant parameters in blood and DNA strand breaks in peripheral leukocytes in healthy volunteers: a randomized controlled trial (ISRCTN68505294). Nutr J 2005;4:33. [64] Collins AR, Duthie SJ, Dobson VL. Direct enzymic detection of endogenous oxidative base damage in human lymphocyte DNA. Carcinogenesis 1993;14:1733–5. [65] Foksinski M, Rozalski R, Guz J, et al. Urinary excretion of DNA repair products correlates with metabolic rates as well as with maximum life spans of different mammalian species. Free Radic Biol Med 2004;37:1449–54. [66] Collins AR. Assays for oxidative stress and antioxidant status: applications to research into the biological effectiveness of polyphenols. Am J Clin Nutr 2005;81:261S–7S. [67] Bowen P, Chen L, Stacewicz-Sapuntzakis M, et al. Tomato sauce supplementation and prostate cancer: lycopene accumulation and modulation of biomarkers of carcinogenesis. Exp Biol Med 2002;227:886–93. [68] Collins AR, Gedik CM, Olmedilla B, Southon S, Bellizzi M. Oxidative DNA damage measured in human lymphocytes: large differences between sexes and between countries, and correlations with heart disease mortality rates. FASEB J 1998;12:1397–400. [69] Moller P, Loft S. Interventions with antioxidants and nutrients in relation to oxidative DNA damage and repair. Mutat Res 2004;551:79–89. [70] Choi SW, Benzie IF, Collins AR, Hannigan BM, Strain JJ. Vitamins C and E: acute interactive effects on biomarkers of antioxidant defence and oxidative stress. Mutat Res 2004;551:109–17. [71] Tsilimigaki S, Messini-Nikolaki N, Kanariou M, Piperakis SM. A study on the effects of seasonal solar radiation on exposed populations. Mutagenesis 2003;18:139–43. [72] Panayiotidis M, Collins AR. Ex vivo assessment of lymphocyte antioxidant status using the comet assay. Free Radic Res 1997;27:533–7. [73] Moller P, Vogel U, Pedersen A, Dragsted LO, Sandstrom B, Loft S. No effect of 600 g fruit and vegetables per day on oxidative DNA damage and repair in healthy human non-smokers. Cancer Epidemiol Biomarkers Prev 2003;12:1016–22. [74] Moller P, Viscovich M, Lykkesfeldt J, Loft S, Jensen A, Poulsen HE. Vitamin C supplementation decreases oxidative DNA damage in mononuclear blood cells of smokers. Eur J Nutr 2004;43:267–74. [75] Giovannelli L, Saieva C, Masala G, et al. Nutritional and lifestyle determinants of DNA oxidative damage: a study in a Mediterranean population. Carcinogenesis 2002;23:1483–9. [76] Gedik CM, Collins A. ESCODD (European Standards Committee on Oxidative DNA Damage). Establishing the background level of base oxidation in human lymphocyte DNA: results of an inter-laboratory validation study. FASEB J 2005;19:82–4. [77] Burger D, Touyz RM. Cellular biomarkers of endothelial health: microparticles, endothelial progenitor cells, and circulating endothelial cells. J Am Soc Hypertens 2012;6(2):85–99. [78] Eguchi M, Monden K, Miwa N. Role of MAPK phosphorylation in cytoprotection by pro-vitamin C against oxidative stress-induced injuries in cultured cardiomyoblasts and perfused rat heart. J Cell Biochem 2003;90:219–26. [79] Nguyen HT, Hsieh MH, Gaborro A, Tinloy B, Phillips C, Adam RM. JNK/SAPK and p38 SAPK-2 mediate mechanical stretch-induced apoptosis via caspase-3 and-9 in NRK-52E renal epithelial cells. Nephron Exp Nephrol 2006;102:49–61. [80] Matsuzawa A, Ichijo H. Redox control of cell fate by MAP kinase: physiological roles of ASK1-MAP kinase pathway in stress signaling. Biochim Biophys Acta Gen Subj 2008;1780:1325–36. [81] Torres M. Mitogen-activated protein kinase pathways in redox signaling. Front Biosci 2003;8:D369–91. [82] Donadelli M, DallaPozza E, Costanzo C, Scupoli MT, Piacentini P, Scarpa A. Increased stability of P21(WAF1/CIP1) mRNA is required for ROS/ERK-dependent pancreatic adenocarcinoma cell growth inhibition by pyrrolidinedithiocarbamate. Biochim Biophys Acta 2006;1763:917–26. [83] Hsu WH, Hsieh YS, Kuo HC, et al. Berberine induces apoptosis in SW620 human colonic carcinoma cells through generation of reactive oxygen species and activation of JNK/p38 MAPK and FasL. Arch Toxicol 2007;81:719–28. [84] Liu Y, Borchert GL, Surazynski A, Hu CA, Phang JM. Proline oxidase activates both intrinsic and extrinsic pathways for apoptosis: the role of ROS/superoxides, NFAT and MEK/ERK signaling. Oncogene 2006;25:5640–7. [85] Barry I. Vitamin C: friends or foe? Nat Rev Cancer 2008;8:830. [86] Liu L, Zhao SP, Gao M, Zhou QCZ, Li YL, Xia B. Vitamin C preserves endothelial function in patients with coronary heart disease after a high-fat meal. Clin Cardiol 2002;25:219–24. [87] Wang Y, Hodge AM, Wluka AE, et al. Effect of antioxidante on knee cartilage and bone in healthy, middle-aged subjects: a cross-sectional study. Arthritis Res Ther 2007;9:1–9. [88] Wintergerst ES, Maggini S, Homig DH. Immune-enhancing role of vitamin C and zinc and effect on clinical conditions. Ann Nutr Metab 2006;50:85–94. [89] Woo A, Kim JH, Jeong YJ, et al. Vitamin C acts indirectly to modulate isotype switching in mouse B cells. Anat Cell Biol 2010;43:25–35.

U

1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 1113 1114 1115 1116 1117 1118 1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133 1134 1135 1136 1137 1138 1139 1140 1141 1142 1143 1144 1145 Q7 1146 1147 1148 Q8 1149 1150 1151 Q9 1152 1153 1154 Q10 1155 1156 Q11 1157 1158 1159 1160 Q12 1161 1162 1163 Q13 1164 1165 1166 1167 1168 1169 1170 1171 1172 1173 1174 1175 1176

13

Please cite this article as: Rajendran P, et al, Antioxidants and human diseases, Clin Chim Acta (2014), http://dx.doi.org/10.1016/j.cca.2014.06.004

1177 1178 1179 1180 1181 1182 Q14 1183 1184 1185 1186 1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200 1201 1202 1203 1204 1205 1206 1207 1208 1209 1210 1211 1212 1213 1214 1215 1216 1217 1218 1219 1220 1221 1222 1223 1224 1225 1226 1227 1228 1229 1230 1231 1232 1233 1234 1235 1236 1237 1238 1239 1240 1241 1242 1243 1244 1245 1246 1247 1248 1249 1250 1251 1252 1253 1254 1255 1256 1257 1258 1259 1260 1261 1262

D

P

R O

O

F

[159] Hunnisett A, Davies S, McLaren-Howard J, Gravett P, Finn M, Gueret-Wardle D. Lipoperoxides as an index of free radical activity in bone marrow transplant recipients. Preliminary observations. Biol Trace Elem Res 1995;47:125–32. [160] Lloyd JV, Davis PS, Emery H, Lander H. Platelet ascorbic acid levels in normal subjects and in disease. J Clin Pathol 1972;25:478–83. [161] Sundström H, Korpela H, Viinikka L, Kauppila A. Serum selenium and glutathione peroxidase, and plasma lipid peroxides in uterine, ovarian or vulvar cancer, and their responses to antioxidants in patients with ovarian cancer. Cancer Lett 1984;24:1–10. [162] Jaakkola K, Lähteenmäki P, Laakso J, Harju E, Tykkä H, Mahlberg K. Treatment with antioxidant and other nutrients in combination with chemotherapy and irradiation in patients with small-cell lung cancer. Anticancer Res 1992;12:599–606. [163] Lockwood K, Moesgaard S, Hanioka T, Folkers K. Apparent partial remission of breast cancer in ‘high risk’ patients supplemented with nutritional antioxidants, essential fatty acids and coenzyme Q10. Mol Aspects Med 1994;15:s231–40. [164] Ladas Elena J, Jacobson Judith S, Kennedy Deborah D, Teel Katherine, Fleischauer Aaron, Kelly Kara M. Antioxidants and cancer therapy: a systematic review. J Clin Oncol 2004;22:517–28. [165] Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature 2001;414:813–20. [166] Maritim AC, Sanders RA, Watkins JB. Diabetes, oxidative stress, and antioxidants: a review. J Biochem Mol Toxicol 2003;17:24–38. [167] Nishigaki I, Rajendran P, Venugopal R, Ekambaram G, Sakthisekaran D, Nishigaki Y. Cytoprotective role of astaxanthin against glycated protein/iron chelate-induced toxicity in human umbilical vein endothelial cells. Phytother Res 2010;24:54–9. [168] Giacco F, Brownlee M. Oxidative stress and diabetic complications. Circ Res 2010;107:1058–70. [169] Avignon A, Hokayem M, Bisbal C, Lambert K. Dietary antioxidants: do they have a role to play in the ongoing fight against abnormal glucose metabolism? Nutrition 2012;28:715–21. [170] Ceriello A, Morocutti A, Mercuri F, et al. Defective intracellular antioxidant enzyme production in type 1 diabetic patients with nephropathy. Diabetes 2000;49:2170–7. [171] DeRubertis FR, Craven PA, Melhem MF. Acceleration of diabetic renal injury in the superoxide dismutase knockout mouse: effects of tempol. Metab Clin Exp 2007;56:1256–64. [172] Hinerfeld D, Traini MD, Weinberger RP, et al. Endogenous mitochondrial oxidative stress: neurodegeneration, proteomic analysis, specific respiratory chain defects, and efficacious antioxidant therapy in superoxide dismutase 2 null mice. J Neurochem 2004;88:657–67. [173] Nishikawa T, Edelstein D, Du XL, et al. Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature 2000;404:787–90. [174] Asaba K, Tojo A, Onozato ML, Goto A, Fujita T. Double-edged action of SOD mimetic in diabetic nephropathy. J Cardiovasc Pharmacol 2007;49:13–9. [175] Mollsten A, Marklund SL, Wessman M, et al. A functional polymorphism in the manganese superoxide dismutase gene and diabetic nephropathy. Diabetes 2007;56:265–9. [176] de Haan JB, Stefanovic N, Nikolic-Paterson D, et al. Kidney expression of glutathione peroxidase-1 is not protective against streptozotocin-induced diabetic nephropathy. Am J Physiol Ren Physiol 2005;289:F544–51. [177] Brezniceanu ML, Liu F, Wei CC, et al. Catalase overexpression attenuates angiotensinogen expression and apoptosis in diabetic mice. Kidney Int 2007;71:912–23. [178] dos Santos KG, Canani LH, Gross JL, Tschiedel B, PiresSouto KE, Roisenberg I. The catalase-262C/T promoter polymorphism and diabetic complications in Caucasians with type 2 diabetes. Dis Markers 2006;22:355–9. [179] Kunisaki M, Bursell SE, Clermont AC, et al. Vitamin-E prevents diabetes-induced abnormal retinal blood-flow via the diacylglycerol-protein kinase-C pathway. Am J Physiol Endocrinol Metab 1995;269:E239–46. [180] Mohora M, Greabu M, Muscurel C, Duţa C, Totan A. The sources and the targets of oxidative stress in the etiology of diabetic complications. Rom J Biophys 2007;17:63–84. [181] Obrosova IG, Fathallah L, Greene DA. Early changes in lipid peroxidation and antioxidativedefense in diabetic rat retina: effect of DL-alpha-lipoic acid. Eur J Pharmacol 2000;398:139–46. [182] Hong JH, Kim MJ, Park MR, et al. Effects on vitamin E on oxidative stress and membrane fluidity brain of streptozotocin-induced diabetic rats. Clin Chim Acta 2004;340:107–15. [183] Coleman MD, Fernandes S, Khanderia L. A preliminary evaluation of a novel method to monitor a triple antioxidant combination (vitamins E, C and alpha-lipoic acid) in diabetic volunteers using in vitro methaemoglobin formation. Environ Toxicol Pharmacol 2003;14:69–75. [184] Panjwani U, Yadav DK, Kumar A, Singh SB, Selvamurthy W. Effect of vitamin C and E supplementation in modulating the peripheral nerve conduction following cold exposure in humans. Int J Biometeorol 2003;48:103–7. [185] Sivan E, Reece EA, Wu YK, Homko CJ, Polansky M, Borenstein M. Dietary vitamin E prophylaxis and diabetic embryopathy: morphologic and biochemical analysis. Am J Obstet Gynecol 1996;175:793–9. [186] Chang TI, Horal M, Jain SK, Wang F, Patel R, Loeken MR. Oxidant regulation of gene expression and neural tube development: insights gained from diabetic pregnancy on molecular causes of neural tube defects. Diabetologia 2003;46:538–45. [187] Otero P, Bonet B, Herrera E, Rabano A. Development of atherosclerosis in the diabetic BALB/c mice—prevention with Vitamin E administration. Atherosclerosis 2005;182:259–65. [188] Hayek T, Kaplan M, Kerry R, Aviram M. Macrophage NADPH oxidase activation, impaired cholesterol fluxes, and increased cholesterol biosynthesis in diabetic mice: a stimulatory role for D-glucose. Atherosclerosis 2007;195:277–86.

N

C

O

R

R

E

C

T

[126] Benelli R, Lorusso G, Albini A, Noonan DM. Cytokines and chemokines as regulators of angiogenesis in health and disease. Curr Pharm Des 2006;12:3101–15. [127] Vendramini-Costa DB, Carvalho JE. Molecular link mechanisms between inflammation and cancer. Curr Pharm Des 2012;18:3831–52. [128] Knaapen AM, Shi TM, Borm PJA, Schins RPF. Soluble metals as well as the insoluble particle fraction are involved in cellular DNA damage induced by particulate matter. Mol Cell Biochem 2002;234:317–26. [129] Hsu TC, Young MR, Cmarik J, Colburn NH. Activator protein 1 (AP-1)- and nuclear factor kappa B (NF-kappa B)-dependent transcriptional events in carcinogenesis. Free Radic Biol Med 2000;28:1338–48. [130] Dhar A, Young MR, Colburn NH. The role of AP-1, NF-kappa B and ROS/NOS in skin carcinogenesis: the JB6 model is predictive. Mol Cell Biochem 2002;234:185–93. [131] Sethi G, Sung B, Aggarwal BB. Nuclear factor-kB activation: from bench to bedside. Exp Biol Med 2008;233:21–31. [132] Lee CH, Jeon YT, Kim SH, Song YS. NF-kappa B as a potential molecular target for cancer therapy. BioFactors 2007;29:19–35. [133] Wang H, Cho CH. Effect of NF-kappa B signaling on apoptosis in chronic inflammation-associated carcinogenesis. Curr Cancer Drug Targets 2010;10:593–9. [134] Kaefer CM, Milner JA. The role of herbs and spices in cancer prevention. J Nutr Biochem 2008;19:347–61. [135] Pool-Zobel B, Veeriah S, Bohmer FD. Modulation of xenobiotic metabolising enzymes by anticarcinogens—focus on glutathione S-transferases and their role as targets of dietary chemoprevention in colorectal carcinogenesis. Mutat Res Fundam Mol Mech Mutagen 2005;591:74–92. [136] Barve A, Khor TO, Nair S, et al. gamma-Tocopherol-enriched mixed tocopherol diet inhibits prostate carcinogenesis in TRAMP mice. Int J Cancer 2009;124:1693–9. [137] Kumaraguruparan R, Seshagiri PB, Hara Y, Nagini S. Chemoprevention of rat mammary carcinogenesis by black tea polyphenols: modulation of xenobioticmetabolizing enzymes, oxidative stress, cell proliferation, apoptosis, and angiogenesis. Mol Carcinog 2007;46:797–806. [138] Lee SB, Kim CY, Lee HJ, Yun JH, Nho CW. Induction of the phase II detoxification enzyme NQO1 in hepatocarcinoma cells by lignans from the fruit of Schisandrachinensis through nuclear accumulation of Nrf2. Planta Med 2009;75:1314–8. [139] Acharya A, Das I, Chandhok D, Saha T. Redox regulation in cancer a double-edged sword with therapeutic potential. Oxidative Med Cell Longev 2010;3:23–34. [140] Huang F, Nie C, Yang Y, et al. Selenite induces redox-dependent Bax activation and apoptosis in colorectal cancer cells. Free Radic Biol Med 2009;46:1186–96. [141] Mebratu Y, Tesfaigzi Y. How ERK1/2 activation controls cell proliferation and cell death is subcellular localization the answer? Cell Cycle 2009;8:1168–75. [142] McCubrey JA, Steelman LS, Abrams SL, et al. Targeting survival cascades induced by activation of Ras/Raf/MEK/ERK, PI3K/PTEN/Akt/mTOR and Jak/STAT pathways for effective leukemia therapy. Leukemia 2008;22:708–22. [143] Eguchi M, Monden K, Miwa N. Role of MAPK phosphorylation in cytoprotection by pro-vitamin C against oxidative stress-induced injuries in cultured cardiomyoblasts and perfused rat heart. J Cell Biochem 2003;90:219–26. [144] Nguyen HT, Hsieh MH, Gaborro A, Tinloy B, Phillips C, Adam RM. JNK/SAPK and p38 SAPK-2 mediate mechanical stretch-induced apoptosis via caspase-3 and -9 in NRK-52E renal epithelial cells. Nephron Exp Nephrol 2006;102:49–61. [145] Matsuzawa A, Ichijo H. Redox control of cell fate by MAP kinase: physiological roles of ASK1-MAP kinase pathway in stress signaling. Biochim Biophys Acta Gen Subj 2008;1780:1325–36. [146] Torres M. Mitogen-activated protein kinase pathways in redox signaling. Front Biosci 2003;8:D369–91. [147] Donadelli M, DallaPozza E, Costanzo C, Scupoli MT, Piacentini P, Scarpa A. Increased stability of P21(WAF1/CIP1) mRNA is required for ROS/ERK-dependent pancreatic adenocarcinoma cell growth inhibition by pyrrolidinedithiocarbamate. Biochim Biophys Acta 2006;1763:917–26. [148] Hsu WH, Hsieh YS, Kuo HC, et al. Berberine induces apoptosis in SW620 human colonic carcinoma cells through generation of reactive oxygen species and activation of JNK/p38 MAPK and FasL. Arch Toxicol 2007;81:719–28. [149] Liu Y, Borchert GL, Surazynski A, Hu CA, Phang JM. Proline oxidase activates both intrinsic and extrinsic pathways for apoptosis: the role of ROS/superoxides, NFAT and MEK/ERK signaling. Oncogene 2006;25:5640–7. [150] Nguyen T, Nioi P, Pickett CB. The Nrf2-antioxidant response element signaling pathway and its activation by oxidative stress. J Biol Chem 2009;284:13291–5. [151] Dhakshinamoorthy S, Jaiswal AK. Small Maf (MafG and MafK) proteins negatively regulate antioxidant response element-mediated expression and antioxidant induction of the NAD(P)H : quinone oxidoreductase1 gene. J Biol Chem 2000;275:40134–41. [152] Li W, Kong AN. Molecular mechanisms of Nrf2-mediated antioxidant response. Mol Carcinog 2009;48:91–104. [153] Kensler TW, Wakabayashi N. Nrf2: friend or foe for chemoprevention? Carcinogenesis 2010;31:90–9. [154] Singh B, Bhat NK, Bhat HK. Induction of NAD(P)H-quinoneoxidoreductase 1 by antioxidants in female ACI rats is associated with decrease in oxidative DNA damage and inhibition of estrogen-induced breast cancer. Carcinogenesis 2012;33:156–63. [155] Lee JS, Surh YJ. Nrf2 as a novel molecular target for chemoprevention. Cancer Lett 2005;224:171–84. [156] Yu X, Kensler T. Nrf2 as a target for cancer chemoprevention. Mutat Res Fundam Mol Mech Mutagen 2005;591:93–102. [157] Clemens MR, Ladner C, Schmidt H, et al. Decreased essential antioxidants and increased lipid hydroperoxides following high-dose radiochemotherapy. Free Radic Res Commun 1989;7:227–32. [158] Clemens MR, Waladkhani AR, Bublitz K, Ehninger G, Gey KF. Supplementation with antioxidants prior to bone marrow transplantation. Wien Klin Wochenschr 1997;109:771–6.

U

1263 1264 1265 1266 1267 1268 1269 1270 1271 1272 1273 1274 1275 1276 1277 1278 1279 1280 1281 1282 1283 1284 1285 1286 1287 1288 1289 1290 1291 1292 1293 1294 1295 1296 1297 1298 1299 1300 1301 1302 1303 1304 1305 1306 1307 1308 1309 1310 1311 1312 1313 1314 1315 1316 1317 1318 1319 1320 1321 1322 1323 1324 1325 1326 1327 1328 1329 1330 1331 1332 1333 1334 1335 1336 1337 1338 1339 1340 1341 1342 1343 1344 1345 1346 1347 1348

P. Rajendran et al. / Clinica Chimica Acta xxx (2014) xxx–xxx

E

14

Please cite this article as: Rajendran P, et al, Antioxidants and human diseases, Clin Chim Acta (2014), http://dx.doi.org/10.1016/j.cca.2014.06.004

1349 1350 1351 1352 1353 1354 Q15 1355 1356 1357 1358 1359 1360 1361 1362 1363 1364 1365 1366 1367 1368 1369 1370 1371 1372 1373 1374 1375 1376 1377 1378 1379 1380 1381 1382 1383 1384 1385 1386 1387 1388 1389 1390 1391 1392 1393 1394 1395 1396 1397 1398 1399 1400 1401 1402 1403 1404 1405 1406 1407 1408 1409 1410 1411 1412 1413 1414 1415 1416 1417 1418 1419 1420 1421 1422 1423 1424 1425 1426 1427 1428 1429 1430 1431 1432 1433 1434

P. Rajendran et al. / Clinica Chimica Acta xxx (2014) xxx–xxx

N C O

R

R

E

C

D

P

R O

O

F

[221] Forgione MA, Weiss N, Heydrick S, et al. Cellular glutathione peroxidase deficiency and endothelial dysfunction. Am J Physiol Heart Circ Physiol 2002;282:H1255–61. [222] Kisucka J, Chauhan AK, Patten IS, et al. Peroxiredoxin1 prevents excessive endothelial activation and early atherosclerosis. Circ Res 2008;103:598–605. [223] Choi MH, Lee IK, Kim GW, et al. Regulation of PDGF signalling and vascular remodelling by peroxiredoxin II. Nature 2005;435:347–53. [224] Park JG, Yoo JY, Jeong SJ, et al. Peroxiredoxin 2 deficiency exacerbates atherosclerosis in apolipoprotein E-deficient mice. Circ Res 2011;109:739–49. [225] Guo X, Yamada S, Tanimoto A, et al. Overexpression of peroxiredoxin 4 attenuates atherosclerosis in apolipoprotein E knockout mice. Antioxid Redox Signal 2012;17:1362–75. [226] Wei Y, Liu XM, Peyton KJ, et al. Hypochlorous acid-induced heme oxygenase-1 gene expression promotes human endothelial cell survival. Am J Physiol Cell Physiol 2009;297:C907–15. [227] Li C, Hossieny P, Wu BJ, Qawasmeh A, Beck K, Stocker R. Pharmacologic induction of heme oxygenase-1. Antioxid Redox Signal 2007;9:2227–39. [228] Gilgun-Sherki Y, Melamed E, Offen D. Oxidative stress induced-neurodegenerative diseases: the need for antioxidants that penetrate the blood brain barrier. Neuropharmacology 2001;40:959–75. [229] Salganik RI. The benefits and hazards of antioxidants: controlling apoptosis and other protective mechanisms in cancer patients and the human population. J Am Coll Nutr 2001;20:464S–72S. [230] Ho YS, Xiong Y, Ma WC, Spector A, Ho DS. Mice lacking catalase develop normally but show differential sensitivity to oxidant tissue injury. J Biol Chem 2004;279:32804–12. [231] Meister A. Mitochondrial changes associated with glutathione deficiency. Biochim Biophys Acta Mol Basis Dis 1995;1271:35–42. [232] DarleyUsmar V, Halliwell B. Blood radicals—reactive nitrogen species, reactive oxygen species, transition metal ions, and the vascular system. Pharm Res 1996;13:649–62. [233] Hansen JM, Go YM, Jones DP. Nuclear and mitochondrial compartmentation of oxidative stress and redox signaling. Annu Rev Pharmacol Toxicol 2006:215–34. [234] Schulz JB, Lindenau J, Seyfried J, Dichgans J. Glutathione, oxidative stress and neurodegeneration. Eur J Biochem 2000;267:4904–11. [235] Sies H. Glutathione and its role in cellular functions. Free Radic Biol Med 1999;27:916–21. [236] Meister A, Anderson ME. Glutathione. Annu Rev Biochem 1983;52:711–60. [237] Sultana R, Butterfield DA. Oxidatively modified GST and MRP1 in Alzheimer's disease brain: implications for accumulation of reactive lipid peroxidation products. Neurochem Res 2004;29:2215–20. [238] Renes J, De Vries EEG, Hooiveld G, Krikken I, Jansen PLM, Muller M. Multidrug resistance protein MRP1 protects against the toxicity of the major lipid peroxidation product 4-hydroxynonenal. Biochem J 2000;350:555–61. [239] Nies AT, Jedlitschky G, Konig J, et al. Expression and immunolocalization of the multidrug resistance proteins, Mrp1–Mrp6 (ABCC1–ABCC6), in human brain. Neuroscience 2004;129:349–60. [240] Conseil G, Deeley RG, Cole SPC. Polymorphisms of MRP1 (ABCC1) and related ATPdependent drug transporters. Pharmacogenet Genomics 2005;15:523–33. [241] Lovell MA, Xie C, Markesbery WR. Decreased glutathione transferase activity in brain and ventricular fluid in Alzheimer's disease. Neurology 1998;51:1562–6. [242] Schipper HM, Bennett DA, Liberman A, et al. Glial heme oxygenase-1 expression in Alzheimer disease and mild cognitive impairment. Neurobiol Aging 2006;27:252–61. [243] Raina AK, Templeton DJ, Deak JC, Perry G, Smith MA. Quinone reductase (NQO1), a sensitive redox indicator, is increased in Alzheimer's disease. Redox Rep 1999;4:23–7. [244] Ramsey CP, Glass CA, Montgomery MB, et al. Expression of Nrf2 in neurodegenerative diseases. J Neuropathol Exp Neurol 2007;66:75–85. [245] Butterfield DA, Drake J, Pocernich C, Castegna A. Evidence of oxidative damage in Alzheimer's disease brain: central role for amyloid beta-peptide. Trends Mol Med 2001;7:548–54. [246] Markesbery WR. Oxidative stress hypothesis in Alzheimer's disease. Free Radic Biol Med 1997;23:134–47. [247] Lovell MA, Xie C, Markesbery WR. Acrolein is increased in Alzheimer's disease brain and is toxic to primary hippocampal cultures. Neurobiol Aging 2001;22:187–94. [248] Markesbery WR, Lovell MA. Four-hydroxynonenal, a product of lipid peroxidation, is increased in the brain in Alzheimer's disease. Neurobiol Aging 1998;19:33–6. [249] Lauderback CM, Hackett JM, Huang FF, et al. The glial glutamate transporter, GLT-1, is oxidatively modified by 4-hydroxy-2-nonenal in the Alzheimer's disease brain: the role of A beta 1-42. J Neurochem 2001;78:413–6. [250] Kanninen K, Malm TM, Jyrkkanen H-K, et al. Nuclear factor erythroid 2-related factor 2 protects against beta amyloid. Mol Cell Neurosci 2008;39:302–13. [251] Resende R, Moreira PI, Proenca T, et al. Brain oxidative stress in a triple-transgenic mouse model of Alzheimer disease. Free Radic Biol Med 2008;44:2051–7. [252] Martinez RCR, Hamani C, de Carvalho MC, et al. Intraoperative dopamine release during globuspallidusinternus stimulation in Parkinson's disease. Mov Disord 2013;28:2027–31. [253] Passamonti L, Salsone M, Toschi N, et al. Dopamine-transporter levels drive striatal responses to apomorphine in Parkinson's disease. Brain Behav 2013;3:249–62. [254] Agid Y, Ruberg M, Javoy-Agid F, et al. Are dopaminergic neurons selectively vulnerable to Parkinson's disease? In: Narabayashi H, Nagatsu T, Yanagisawa N, Mizuno Y, editors. Advances in Neurology; Parkinson's Disease: From basic research to treatment; 1993. p. 148–64. [255] Smeyne M, Smeyne RJ. Glutathione metabolism and Parkinson's disease. Free Radic Biol Med 2013;62:13–25. [256] Smith TS, Parker WD, Bennett JP. L-dopa increases nigral production of hydroxyl radicals in-vivo—potential l-dopa toxicity. Neuroreport 1994;5:1009–11. [257] Wick MM, Byers L, Frei E. L-dopa—selective toxicity for melanoma cells invitro. Science 1977;197:468–9.

E

T

[189] Reaven PD, Herold DA, Barnett J, Edelman S. Effects of vitamin-E on susceptibility of low-density-lipoprotein and low-density lipoprotein subfractions to oxidation and on protein glycation in niddm. Diabetes Care 1995;18:807–16. [190] Salonen JT, Ylaherttuala S, Yamamoto R, et al. Autoantibody against oxidized LDL and progression of carotid atherosclerosis. Lancet 1992;339:883–7. [191] Bantle JP, Wylie-Rosett J, Albright AL, et al. Nutrition recommendations and interventions for diabetes: a position statement of the American Diabetes Association. Diabetes Care 2008;31(1):S61–78. [192] Landmesser U, Drexler H. General concepts about oxidative stress. Dev Cardiovasc Med 2006;258:1–15. [193] Ohara Y, Peterson TE, Harrison DG. Hypercholesterolemia increases endothelial superoxide anion production. J Clin Invest 1993;91:2546–51. [194] Hink U, Li HG, Mollnau H, et al. Mechanisms underlying endothelial dysfunction in diabetes mellitus. Circ Res 2001;88:E14–22. [195] Victor VM, Rocha M, Sola E, Banuls C, Garcia-Malpartida K, Hernandez-Mijares A. Oxidative stress, endothelial dysfunction and atherosclerosis. Curr Pharm Des 2009;15:2988–3002. [196] Sugamura K, Keaney Jr JF. Reactive oxygen species in cardiovascular disease. Free Radic Biol Med 2011;51:978–92. [197] Libby P, Ridker PM, Hansson GK. Leducq Transatlantic Network A. Inflammation in atherosclerosis from pathophysiology to practice. J Am Coll Cardiol 2009;54:2129–38. [198] Fuhrman B, Volkova N, Aviram M. Oxidative stress increases the expression of the CD36 scavenger receptor and the cellular uptake of oxidized low-density lipoprotein in macrophages from atherosclerotic mice: protective role of antioxidants and of paraoxonase. Atherosclerosis 2002;161:307–16. [199] Shirpoor A, Ansari M-HK, Heshmatian B, et al. Decreased blood pressure with a corresponding decrease in adhesive molecules in diabetic rats caused by vitamin E administration. J Diabetes 2012;4:362–8. [200] Saeed RW, Peng T, Metz CN. Ascorbic acid blocks the growth inhibitory effect of tumor necrosis factor-alpha on endothelial cells. Exp Biol Med 2003;228:855–65. [201] Rossig L, Hoffmann J, Hugel B, et al. Vitamin C inhibits endothelial cell apoptosis in congestive heart failure. Circulation 2001;104:2182–7. [202] Libby P, Aikawa M. Vitamin C, collagen, and cracks in the plaque. Circulation 2002;105:1396–8. [203] Axelrad TW, Deo DD, Ottino P, et al. Platelet-activating factor (PAF) induces activation of matrix metalloproteinase 2 activity and vascular endothelial cell invasion and migration. FASEB J 2004;18:568–70. [204] Yoshikawa K, Takahashi S, Imamura Y, Sado Y, Hayashi T. Secretion of non-helical collagenous polypeptides of alpha 1(IV) and alpha 2(IV) chains upon depletion of ascorbate by cultured human cells. J Biochem 2001;129:929–36. [205] Kawashima S, Yokoyama M. Dysfunction of endothelial nitric oxide synthase and atherosclerosis. Arterioscler Thromb Vasc Biol 2004;24:998–1005. [206] Heller R, Unbehaun A, Schellenberg B, Mayer B, Werner-Felmayer G, Werner ER. Lascorbic acid potentiates endothelial nitric oxide synthesis via a chemical stabilization of tetrahydrobiopterin. J Biol Chem 2001;276:40–7. [207] Gao F, Yao CL, Gao EH, et al. Enhancement of glutathione cardioprotection by ascorbic acid in myocardial reperfusion injury. J Pharmacol Exp Ther 2002;301:543–50. [208] Vink H, Constantinescu AA, Spaan JAE. Oxidized lipoproteins degrade the endothelial surface layer—implications for platelet-endothelial cell adhesion. Circulation 2000;101:1500–2. [209] Fukai T, Siegfried MR, Ushio-Fukai M, Cheng Y, Kojda G, Harrison DG. Regulation of the vascular extracellular superoxide dismutase by nitric oxide and exercise training. J Clin Invest 2000;105:1631–9. [210] Narang D, Sood S, Thomas MK, Dinda AK, Maulik SK. Effect of dietary palm olein oil on oxidative stress associated with ischemic-reperfusion injury in isolated rat heart. BMC Pharmacol 2004;4:29. [211] Das S, Powell SR, Wang P, et al. Cardioprotection with palm tocotrienol: antioxidant activity of tocotrienol is linked with its ability to stabilize proteasomes. Am J Physiol Heart Circ Physiol 2005;289:H361–7. [212] Carlson D, Maass DL, White DJ, Tan J, Horton JW. Antioxidant vitamin therapy alters sepsis-related apoptotic myocardial activity and inflammatory responses. Am J Physiol Heart Circ Physiol 2006;291:H2779–89. [213] Tian N, Moore RS, Braddy S, et al. Interactions between oxidative stress and inflammation in salt-sensitive hypertension. Am J Physiol Heart Circ Physiol 2007;293: H3388–95. [214] Papparella I, Ceolotto G, Berto L, et al. Vitamin C prevents zidovudine-induced NAD(P) H oxidase activation and hypertension in the rat. Cardiovasc Res 2007;73:432–8. [215] Matejikova J, Kucharska J, Pancza D, Ravingerova T. The effect of antioxidant treatment and NOS inhibition on the incidence of ischemia-induced arrhythmias in the diabetic rat heart. Physiol Res/Acad Sci Bohem 2008;57:S55–60. [216] Chu Y, Iida S, Lund DD, et al. Gene transfer of extracellular superoxide dismutase reduces arterial pressure in spontaneously hypertensive rats: role of heparinbinding domain. Circ Res 2003;92:461–8. [217] Sentman ML, Brannstrom T, Westerlund S, et al. Extracellular superoxide dismutase deficiency and atherosclerosis in mice. Arterioscler Thromb Vasc Biol 2001;21:1477–82. [218] Yang H, Roberts LJ, Shi MJ, et al. Retardation of atherosclerosis by overexpression of catalase or both Cu/Zn-superoxide dismutase and catalase in mice lacking apolipoprotein E. Circ Res 2004;95:1075–81. [219] van Empel VP, Bertrand AT, van Oort RJ, et al. EUK-8, a superoxide dismutase and catalase mimetic, reduces cardiac oxidative stress and ameliorates pressure overload-induced heart failure in the harlequin mouse mutant. J Am Coll Cardiol 2006;48:824–32. [220] Blankenberg S, Rupprecht HJ, Bickel C, et al. Glutathione peroxidase 1 activity and cardiovascular events in patients with coronary artery disease. N Engl J Med 2003;349:1605–13.

U

1435 1436 1437 1438 1439 1440 1441 1442 1443 1444 1445 1446 1447 1448 1449 1450 1451 1452 1453 1454 1455 1456 1457 1458 1459 1460 1461 1462 1463 1464 1465 1466 1467 1468 1469 1470 1471 1472 1473 1474 1475 1476 1477 1478 1479 1480 1481 1482 1483 1484 1485 1486 1487 1488 1489 1490 1491 1492 1493 1494 1495 1496 1497 1498 1499 1500 1501 1502 1503 1504 1505 1506 1507 1508 1509 1510 1511 1512 1513 1514 1515 1516 1517 1518 1519 1520

15

Please cite this article as: Rajendran P, et al, Antioxidants and human diseases, Clin Chim Acta (2014), http://dx.doi.org/10.1016/j.cca.2014.06.004

1521 1522 1523 1524 1525 1526 1527 1528 1529 1530 1531 1532 1533 1534 1535 1536 1537 1538 1539 1540 1541 1542 1543 1544 1545 1546 1547 1548 1549 1550 1551 1552 1553 1554 1555 1556 1557 1558 1559 1560 1561 1562 1563 1564 1565 1566 1567 1568 1569 1570 1571 1572 1573 1574 1575 1576 1577 1578 1579 1580 1581 1582 1583 1584 1585 1586 1587 1588 1589 1590 1591 1592 1593 1594 1595 1596 1597 1598 1599 1600 1601 1602 1603 1604 1605 1606

16

[263] Rajendran P, Li F, Manu KA, et al. Gamma-Tocotrienol is a novel inhibitor of constitutive and inducible STAT3 signalling pathway in human hepatocellular carcinoma: potential role as an antiproliferative, pro-apoptotic and chemosensitizing agent. Br J Pharmacol 2011;163:283–98. [264] Rengarajan T, Rajendran P, Nandakumar N, Balasubramanian MP, Nishigaki I. Cancer preventive efficacy of marine carotenoid fucoxanthin: cell cycle arrest and apoptosis. Nutrients 2013;5:4978–89. [265] Nandakumar N, Rengarajan T, Balamurugan A, Balasubramanian MP. Modulating effects of hesperidin on key carbohydrate-metabolizing enzymes, lipid profile, and mbrane-bound adenosine triphosphatases against 7,12-dimethylbenz(a)anthracene-induced breast carcinogenesis. Hum Exp Toxicol 2013. http://dx.doi.org/ 10.1177/0960327113485252.

N

C

O

R

R

E

C

T

E

D

P

R O

O

F

[258] Hunot S, Brugg B, Ricard D, et al. Nuclear translocation of NF-kappa B is increased in dopaminergic neurons of patients with Parkinson disease. Proc Natl Acad Sci U S A 1997;94:7531–6. [259] Blum D, Torch S, Nissou MF, Verna JM. 6-Hydroxydopamine-induced nuclear factor-kappaB activation in PC12 cells. Biochem Pharmacol 2001;62:473–81. [260] Levites Y, Youdim MBH, Maor G, Mandel S. Attenuation of 6-hydroxydopamine (6-OHDA)-induced nuclear factor-kappaB (NF-kappa B) activation and cell death by tea extracts in neuronal cultures. Biochem Pharmacol 2002;63:21–9. [261] Rajendran P, Ekambaram G, Sakthisekaran D. Cytoprotective effect of mangiferin on benzo(a)pyrene-induced lung carcinogenesis in Swiss albino mice. Basic Clin Pharmacol Toxicol 2008;103:137–42. [262] Ganapathy E, Peramaiyan R, Rajasekaran D, Venkataraman M, Dhanapal S. Modulatory effect of naringenin on N-methyl-N′-nitro-N-nitrosoguanidine- and saturated sodium chloride-induced gastric carcinogenesis in male Wistar rats. Clin Exp Pharmacol Physiol 2008;35:1190–6.

U

1607 1608 1609 1610 1611 1612 1613 1614 1615 1616 1617 1618 1619 1620 1621 1634

P. Rajendran et al. / Clinica Chimica Acta xxx (2014) xxx–xxx

Please cite this article as: Rajendran P, et al, Antioxidants and human diseases, Clin Chim Acta (2014), http://dx.doi.org/10.1016/j.cca.2014.06.004

1622 1623 1624 1625 1626 1627 1628 1629 1630 1631 1632 1633

Antioxidants and human diseases.

Oxidative stress plays a pivotal role in the development of human diseases. Reactive oxygen species (ROS) that includes hydrogen peroxide, hyphochloru...
1MB Sizes 5 Downloads 3 Views