2023 The comparison of antioxidant properties and nutrigenomic redox-related

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Free Radical Biology and Medicine 209 (2023) 239–251

Contents lists available at ScienceDirect

Free Radical Biology and Medicine


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

The comparison of antioxidant properties and nutrigenomic redox-related


activities of vitamin C, C-vitamers, and other common ascorbic
acid derivatives
Patrycja Jakubek a, b, *, Klaudia Suliborska a, Monika Kuczyńska a, Muhammad Asaduzzaman a,
Karol Parchem a, Izabela Koss-Mikołajczyk a, Barbara Kusznierewicz a, Wojciech Chrzanowski a,
Jacek Namieśnik a, Agnieszka Bartoszek a
a
Faculty of Chemistry, Gdańsk University of Technology, 80-233, Gdansk, Poland
b
Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093, Warsaw, Poland

A R T I C L E I N F O A B S T R A C T

Keywords: The term ‘vitamin C’ describes a group of compounds with antiscorbutic activity of L-ascorbic acid (AA). Despite
Antioxidant activity AA’s omnipresence in plant-derived foods, its derivatives have also been successfully implemented in the food
Redox homeostasis industry as antioxidants, including the D-isomers, which lack vitamin C activity. This study aimed to determine
Oxidative damage
the relationship between redox-related activities for five derivatives of AA using electrochemical, chemical, and
Gene expression
Essential nutrients
biological approaches. Here we report that AA, C-vitamers, and other commonly consumed AA derivatives differ
in their redox-related activities. As long as the physiological range of concentrations was maintained, there was
no simple relationship between their redox properties and biological activity. Clear distinctions in antioxidant
activity were observed mostly at high concentrations, which were strongly correlated with electrochemical and
kinetic parameters describing redox-related properties of the studied compounds. Despite obvious similarities in
chemical structures and antioxidant activity, we showed that C-vitamers may exhibit different nutrigenomic
effects. Together, our findings provide a deeper insight into so far underinvestigated area combining chemical
properties with biological activities of commonly applied AA derivatives.

1. Introduction high as 3 g administered 6 times a day resulted in the peak plasma


concentrations not exceeding 220 μM [3]. In tissues, ascorbate accu­
The term essential nutrients refers to substances that are required for mulates in millimolar concentrations that are not possible to be reached
the proper physiological function of human organism, but cannot be in plasma unless pharmacological doses are provided intravenously.
produced endogenously – either at all or in sufficient amounts – and Except for plant-derived sources of vitamin C, AA derivatives can be also
thus, must be provided daily with food [1]. Among such dietary com­ consumed as food additives. The group of ascorbates used in the food
ponents, some display antioxidant activity, e.g., vitamin C. The term industry includes, among others, L-ascorbic acid (E300, AA), sodium
“vitamin C” describes a group of L-ascorbic acid (AA) derivatives, which L-ascorbate (E301, NaA), and calcium L-ascorbate (E302, CaA), which
possess the biological activity of vitamin C. Such structurally related exhibit the biological function of vitamin C, as well as D-isoascorbic acid
compounds can be also named vitamers, or in this particular case – (E315, iAA, also known as erythorbic acid) and sodium D-isoascorbate
C-vitamers [1]. Vitamin C is widely distributed in foods, mostly of plant (E316, iNaA, also known as sodium erythorbate), which lack biological
origin, and its high content was reported in black currants, peppers, kiwi activity of vitamin C (Fig. 1a). The enrichment of food products in AA
fruits, papayas, and oranges [2]. The dietary intake of vitamin C de­ and its derivatives increases their nutritional and sensory value, inhibits
termines its plasma concentration, which reaches 40–80 μM in healthy browning, stabilizes flavour and colour as well as protects against po­
individuals consuming several servings of fruits and vegetables daily. In tential spoilage and oxidation of other food components [4]. Besides,
the case of oral supplementation of AA by healthy volunteers, doses as different C-vitamers can be also consumed in the form of dietary

* Corresponding author. Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., 02-093, Warsaw, Poland.
E-mail address: p.jakubek@nencki.edu.pl (P. Jakubek).

https://doi.org/10.1016/j.freeradbiomed.2023.10.400
Received 11 September 2023; Received in revised form 17 October 2023; Accepted 19 October 2023
Available online 20 October 2023
0891-5849/© 2023 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
P. Jakubek et al. Free Radical Biology and Medicine 209 (2023) 239–251

supplements with recommended supplementation doses, which often 6H2O and Na2S2O3 × 5H2O, which were dissolved in 1 M HCl. Spec­
exceed the recommended dietary allowance of vitamin C by several-fold. trophotometric tests were conducted with DPPH (1-diphenyl-2-picrylhy­
Based on the available evidence, the European Food Safety Authority drazyl) and ABTS (2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic
has conducted thorough evaluations and determined that the con­ acid) diammonium salt) radicals from Sigma Aldrich (USA). Their stock
sumption of AA and its derivatives does not pose any health risks, thus solutions were prepared using analytical grade methanol purchased
there is no need to define an acceptable daily intake for these com­ from Avantor Performance Materials (Poland). ABTS and DPPH solu­
pounds [5–7]. tions were prepared in analytical grade methanol, as discussed in detail
Vitamin C plays multiple essential roles in the human organism, most in the response for Reviewers and indicated in Method description in
of which stem from its ability to donate electrons, which makes it a Section 2.3. Acetone and ethanol were used in other procedures (as
physiologically essential reducing agent [8]. In light of this, it is some­ mentioned in the next sentence), but not for stock solutions of ABTS and
what surprising that to date no complex evaluation of the (electro) DPPH. Fresh free-range hen eggs obtained from a local Polish eco-farm
chemical properties as well as antioxidant and nutrigenomic activities of to avoid pesticide contamination were used for the preparation of li­
C-vitamers and other AA derivatives has been performed. Therefore, in posomes. Reagents applied for the preparation of liposomes included
this study, we aimed to determine the relationship between chemically acetone, methanol, ethanol (Avantor Performance Materials, Poland),
defined antioxidant properties of vitamin C, C-vitamers, and other and hexane (Merck, USA), all of analytical grade. Hemoglobin isolated
common AA derivatives and their impact on redox homeostasis in a from bovine blood (in the form of lyophilized powder), 4-(2-hydrox­
cellular setting mimicking the human alimentary tract. To the best of our yethyl)-1-piperazineethanesulfonic acid (HEPES) (≥99.5%) from Sigma
knowledge, such a comprehensive investigation combining chemistry Aldrich (USA), and nitrogen (99.996%) from Oxygen S.c. (Poland) were
with the biological effects of five different AA derivatives, going beyond used for monitoring lipid peroxidation. The Protein Carbonyl content assay
food processing applications, has not been performed so far. The kit, bovine serum albumin (BSA), and Bicinchoninic acid (BCA) kit were
chemical assessment of antioxidant activity was conducted by applying purchased from Sigma Aldrich (USA) and used for the determination of
electrochemical techniques such as potentiometric titration (PT) and the inhibition of protein oxidation. All reagents for cell culture such as
differential pulse voltammetry (DPV) as well as simple spectrophoto­ phosphate-buffered saline (PBS), McCoy’s 5A medium, trypsin, FBS, and
metric tests employing ABTS and DPPH radicals. With the use of the antibiotics (penicillin and streptomycin) were purchased from Sigma
ABTS and DPPH tests, we established stoichiometric n10 values that Aldrich (USA). For the MTT test, thiazolyl blue tetrazolium bromide
reflect the number of radical molecules that can be scavenged by one (MTT) and dimethyl sulfoxide (DMSO) from Sigma-Aldrich (USA) were
molecule of an antioxidant within a fixed predefined (10 min) reaction used. The CAA assay was carried out using the OxiSelect Cellular Anti­
time. Next, we determined the ability of AA derivatives to modulate oxidant Assay kit purchased from Cell Biolabs, Inc. (USA), and Hanks’
protein and lipid oxidation kinetics using another cell-free system balanced salt solution (HBSS) from Sigma Aldrich (USA). A hydrogen
mimicking intestinal conditions. peroxide solution (30% v/v) as well as all ingredients used for the
In the biological part of this study, human colon adenocarcinoma preparation of buffers for comet assay were purchased from Sigma
HT29 cell line was used as a cellular model representing human intes­ Aldrich (USA) unless stated otherwise. For RNA isolation and assessment
tinal epithelium that is directly exposed to ingested food. When cultured of gene expression, the RNeasy Mini Kit, RNase-Free DNase set, QIAsh­
under standard growth conditions (25 mM glucose, 10% fetal bovine redders, RT2 First Strand Kit, RT2 SybrGreen qPCR Mastermix, and RT2
serum (FBS)), HT29 cells exhibit the phenotype of undifferentiated Profiler PCR Arrays for Oxidative Stress (PAHS 065Z) were provided by
colonic epithelial cells [9]. Moreover, the proteomic analysis of cell lines Qiagen (Germany).
and scrapings of the human intestinal epithelium showed that HT29
cells express proteins that are characteristic of human intestinal 2.2. Electrochemical measurements
epithelium in vivo [10]. As such, on one hand, they resemble intestinal
cells, and on the other hand, their metabolism is characteristic of the 2.2.1. Standard reduction potential by potentiometric titration
metabolism of cancer cells, thus they are more sensitive to changes in The PT and mathematical analysis of experimental titration curves
redox environment compared to normal cells [11]. We needed a model were performed as described before [20]. Briefly, PT was performed
resembling human alimentary tract but at the same time enabling the using JENCO 6230 N equipment (China) and platinum measuring
detection of even very slight differences in response to antioxidant electrode (Micro Combination Redox Electrode, Ag|AgCl inner reference
treatments. The experimental design involved the assessment of cyto­ electrode (RE), JENCO, China). The titration was carried out using a
toxicity (MTT test), cellular antioxidant activity (CAA assay), genotox­ solution of K3[Fe(CN)6]. The tested compounds were dissolved at a
icity, and DNA protection against oxidative damage (comet assay) as concentration of 1 mg/mL using 0.01 M PBS (pH = 7.4 ± 0.1), except for
well as microarray analysis of changes in the expression of 84 CaA. The latter compound was dissolved in ultra-pure water due to its
redox-related genes. The results of this study shine some new light on low solubility in phosphate buffers. The solution of CaA had a pH equal
antioxidant activity regarding complex relationships between chemical to 3.83 ± 0.01, which was measured at the equivalence point of the
structures, electrochemistry as well as biological activity at both cellular titration curve; this value was further used in calculations. The mea­
and nutrigenomic levels. surements were carried out at 37 ◦ C (± 0.1 ◦ C). Finally, the titration
curves E = f(Vtitr.) were analyzed using the sigmoidal 5-parameter model
2. Materials and methods [20] by SigmaPlot 13.0 software (Systat Software Inc., UK). The po­
tential at the equivalence point was read directly from the mathematical
2.1. Chemicals and reagents model and subsequently, the standard reduction potential (E0) of a given
redox couple was calculated. The accurate potential of RE (ϵ) was
AA and its derivatives investigated in this study included: L- measured by the titration of FeCl3 × 6H2O and Na2S2O3 × 5H2O, whose
(+)-ascorbic acid (AA), D-(− )-isoascorbic acid (iAA), sodium L- E0 values are known [20,21].
(+)-ascorbate (NaA), sodium D-(− )-isoascorbate (iNaA) and calcium L-
(+)-ascorbate (CaA), which were purchased from Sigma-Aldrich (USA). 2.2.2. Antioxidant power by differential pulse voltammetry
Hexacyanoferrate(III) potassium (K3[Fe(CN)6]), iron(III) chloride The DPV was performed as described previously [21] with minor
hexahydrate (FeCl3 × 6H2O), sodium thiosulfate pentahydrate modifications. Briefly, 6 mM stock solutions of the studied compounds
(Na2S2O3 × 5H2O) were purchased from Sigma Aldrich (USA) and used were prepared using 0.1 M sodium phosphate buffer (pH = 7.4 ± 0.1) as
in potentiometric titration experiments. All solutions were prepared using a solvent, except for CaA, whose solution was prepared in 0.1 M KCl (pH
purified water (QPLUS185 system, Millipore, USA), except for FeCl3 × = 7.0 ± 0.1). The DPV was carried out using Gamry Reference 600

240
P. Jakubek et al. Free Radical Biology and Medicine 209 (2023) 239–251

potentiostat (Gamry Instruments, Inc., USA). The three-electrode system obtained liposomes were diluted with HEPES buffer (10 mM, pH 7.2) to
consisting of a glassy carbon electrode (BASi, USA) (working electrode the final PL concentration of 0.2 mg/mL. Then, the liposomes were
(WE)), platinum wire (counter electrode (CE)), and the Ag|AgCl elec­ sonicated again as described above to obtain small unilamellar vesicles
trode (reference electrode, RE) from Hydromet S.c. (Poland) was (SUVs). The liposome suspension was filtered on a 0.2 μm Acrodisc sy­
applied. The area of WE (AWE) was equal to 0.162 cm2, which was ringe filter with a nylon membrane to remove potential titanium parti­
determined by cyclic voltammetry of 1 mM K3[Fe(CN)]6 solution in 0.1 cles [13]. The liposome size distribution and dispersity (polydispersity
M KCl. The DPV measurements were recorded in the potential range of index, PDI) were determined using a non-invasive black scatter light
− 0.3 to +0.9 V. The other settings included the potential scan rate (v) of method (NIBS) [14]. For this purpose, a Zetasizer Nano ZS (Malvern,
0.1 V s− 1, the pulse height of 0.05 V, the pulse time of 0.1 s, and the United Kingdom) equipped with helium-neon laser diffraction operating
sample period of 0.5 s. After every measurement, the surface of WE was at 633 nm was employed. All measurements were performed at 37 ◦ C.
polished by alumina suspension (Buehler, USA) on the microcloth pads The obtained PL fraction was used to observe the potential modulation
(BASi, USA) and then rinsed with ultra-pure water and methanol. The of lipid peroxidation and protein carbonylation processes by AA
electrochemical cell was cleaned with a solution of 95% H2SO4 with derivatives.
0.002% of KMnO4 (w/v).
The value of antioxidant power (AOP) was calculated using Equation 2.4.2. Modulation of lipid peroxidation
(1) and was expressed in W•cm− 2: The kinetics of lipid peroxidation was assessed by monitoring the
formation of conjugated dienes in the structure of unsaturated fatty
AOE
AOP = (1) acids (UFAs) build-in PL molecules. For this purpose, liposome suspen­
tf − ti
sion (0.2 mg/mL) was placed in a quartz cell with a 1 cm path length and
where: tf-ti – the difference between the time of the starting point of an incubated in the spectrophotometer thermostat at 37 ◦ C for 7 min. Then,
aqueous solutions of AA or its derivatives were added to obtain a con­
oxidation peak (ti) and its end (tf) [s], AOE – a specific parameter of
antioxidant energy [J × cm− 2] calculated according to Equation (2): centration in the range of 10–100 μM and the mixture was incubated for
a further 2 min. The liposome oxidation was initiated by the addition of
1 ∑
f
an aqueous solution of hemoglobin in the final concentration of 2.5 μM
AOE = − (E − ε) • I • dt (2) (10 μM based on haem iron), which was considered as t = 0 min. The
AWE i
kinetics of oxidation was examined by monitoring the change of ab­
where: E − an applied potential vs. RE [V], ε – a correction term (taking sorption at the wavelength of 234 nm at 37 ◦ C for 120 min at 1 min
into consideration the liquid junction between the WE and RE, identified intervals using a NanoDrop 2000c spectrophotometer (Thermo Scienti­
in the same way as described in Section 2.2.1), I – current measured in fic, USA). The oxidation rates were determined as the slope of the
relation to the background [A], t – potential sampling time [dt = 0.5 s]. regression line for the linear range of absorbance at 234 nm versus re­
action time. All experiments were carried out in three independent
repetitions.
2.3. Antioxidant activity by spectrophotometric tests
2.4.3. Modulation of protein carbonylation
The colorimetric determination of the antioxidant activity of inves­ The effect of AA and its derivatives on protein oxidation was assessed
tigated compounds in a cell-free system and subsequent analysis of ob­ by quantification of the formed carbonyl groups (aldehyde and ketone)
tained results were carried out as described previously [20]. Briefly, the in the structure of a model protein, i.e., BSA. For this purpose, the
absorbance of the methanol solution of ABTS or DPPH radical was Protein Carbonyl content assay kit (Sigma Aldrich) was used following
adjusted to 0.8 ± 0.05 at 734 nm and 0.9 ± 0.05 at 515 nm, respectively. the protocol available on the manufacturer’s website (https://www.
Stock solutions of tested compounds were prepared in 96% ethanol and sigmaaldrich.com/deepweb/assets/sigmaaldrich/product/documents
diluted (using the same solvent) to concentrations falling within a linear /263/399/mak094bul-mk.pdf). BSA solution in HEPES buffer (10 mM,
range of the assay. Measurements of absorbance were performed using a pH 7.4) was mixed with liposomes (prepared as described in section
TECAN Infinite M200 spectrophotometer (Tecan Group Ltd., 2.4.1) to obtain the final BSA and PL concentrations of 0.5 mg/mL each.
Switzerland) after 10 min of incubation at 25 ◦ C, 37 ◦ C, or 41 ◦ C. The Then, the mixture was sonicated as already described to obtain SUVs.
results of antioxidant activity determinations were calculated into Finally, to initiate the protein oxidation, 2 mL of BSA/PL mixture was
stoichiometry value (n10) as described previously [20]. incubated with 0.34 mg/mL of hemoglobin and different concentrations
of tested compounds (10–1000 μM) or HEPES buffer (control) for 4 h at
2.4. Kinetics of protein and lipid oxidation 37 ◦ C. After the incubation, proteins were precipitated from the mixture
using 50 μL of 100% trichloroacetic acid (TCA) solution, vortexed, left
2.4.1. Preparation of liposomes on ice for 5 min, and then centrifuged at 13,000 × g for 5 min. Further
The phospholipid (PL) fraction from hen egg yolk was isolated using steps were performed as described in the manufacturer’s protocol.
methanol followed by PL precipitation in acetone cooled to − 20 ◦ C, as Briefly, protein suspension was mixed with 2,4-dinitrophenylhydrazine
described previously [12]. PL fraction was dissolved in a mixture of (DNPH), and the samples were incubated at room temperature for 10
chloroform/methanol (2:1 v/v) to reach the concentration of 5 mg/mL. min. Afterwards, 100% TCA was used to precipitate proteins, the sam­
Then, the solvent mixture was evaporated under reduced pressure using ples were left on ice for another 5 min and subsequently centrifuged. The
a rotary evaporator at a temperature below 40 ◦ C. The solvent residue supernatant was removed, and the remaining pellet was suspended in
was removed with a gentle stream of nitrogen for 30 min. The resulting ice-cold acetone, treated with ultrasounds, and centrifuged. The samples
thin lipid film was hydrated at room temperature in HEPES buffer (10 were washed twice with acetone to remove unbound DNPH. After the
mM, pH 7.2). For this purpose, the buffer was added to the round bottom second wash, the pellet was dissolved in 6 M guanidine solution, and
flask containing lipid film, and the content was stirred in the nitrogen 100 μL of the obtained solution was transferred to the 96-well plate (in
atmosphere using an orbital shaker at 37 ◦ C for 30 min. The obtained PL three repetitions per each sample). The measurement of absorbance was
vesicles (1 mg/mL) were then sonicated at the ambient temperature (8 carried out at 375 nm using a TECAN Infinite M200 plate reader. The
min, 20 kHz) using Vibra-Cell VCX 750 ultrasound sources fitted with a protein oxidation was expressed as a content of carbonylated proteins.
3 mm diameter titanium micro-tip (Sonics, USA). Power delivery was The amount of carbonylated proteins in a sample per well was calculated
controlled as an amplitude percentage that was set at 40%. A pulsed from Equation (3):
duty cycle of 30 s on and 10 s off was used for all experiments. The

241
P. Jakubek et al. Free Radical Biology and Medicine 209 (2023) 239–251

A375 1000 μM. The cells treated with solvent only served as negative controls,
Ccp = •V (3)
εmM • l whereas cells treated with 200 μM hydrogen peroxide for 1 h served as a
positive control. For protection against DNA damage tests, the treatment
where: Ccp – the content of carbonylated proteins in a well [nmol], A375 conditions were modified as follows: right after the 24 h treatment with
– the absorbance at 375 nm, ƐmM – the millimolar extinction coefficient tested antioxidants at final concentrations of 50, 100, or 200 μM was
1
(22 mM M•cm ), l – an optical path in a well of a 96-well plate (0.2893 cm), finished, the medium was removed and HT29 cells were subjected to
V – the total reaction volume in a well (100 μL). additional 1 h exposure under cell culture conditions to 150 μM
To determine the amount of protein per sample, 5 μL of protein hydrogen peroxide solution prepared in fresh medium. Afterwards,
suspension was transferred to another 96-well transparent plate and comet assay was performed as described previously [17]. The cell lysis
BCA assay was performed. The amount of carbonylated proteins per mg was carried out overnight, while DNA unwinding in electrophoretic
of protein was calculated using Equation (4): buffer lasted 20 min and was followed by 30 min of electrophoresis (26
Ccp V, 300 mA) in darkness. DNA was stained with cold SybrGreen solution
CP = • 1, 000 • D (4) for 30 min and washed with cold ultra-pure water for another 5 min. The
P
stained nuclei were examined under a fluorescence microscope (Zeiss,
where: CP – the total content of carbonylated proteins [nmol/mg pro­ ImagerZ2, USA) and scanned using the Metafer4 scanning system
tein], P – the amount of protein from standard well x 20 [μg/well], 1000 (Germany). For genotoxicity testing, the analysis was performed
– conversion factor (from μg to mg), D – sample dilution. employing the Metafer4 system and involved counting 200 consecutive
nuclei per gel. For testing protection against DNA damage, the analysis
2.5. Cell culture was performed with Comet Score 2.0 software and involved counting
100 consecutive nuclei per gel. The results of comet assay were
Human colon adenocarcinoma HT29 cells were provided and expressed as %DNA in the comet tail.
authenticated by the American Type Culture Collection (ATCC, USA).
The cells were cultured in McCoy’s medium supplemented with 10% 2.6. Microarray analysis
FBS and penicillin/streptomycin and grown in a humidified atmosphere
(37 ◦ C, 5% CO2) in a Smart cell incubator (Heal Force, China). Cultured HT29 cells were seeded in 24-well tissue culture plates at a density of
cells were regularly checked for mycoplasma contamination using 100,000 cells per well and were allowed to settle for 24 h at 37 ◦ C. Then,
Universal Mycoplasma Detection Kit from ATCC (USA). The solutions of the cells were treated with AA and CaA at 50 and 200 μM concentrations
antioxidants used in cell-based tests were additionally sterilized by for 24 h (37 ◦ C, 5% CO2). The tested compounds were dissolved in ultra-
passing through Millex sterile R33 mm (0.22 μm) syringe-driven filters pure water and sterilized with the aid of syringe filters (0.22 μm). Total
(Millipore, USA) to avoid contamination. RNA isolation was performed with the use of the RNeasy Mini kit pro­
tocol with a homogenization step including the use of QIAshredders
2.5.1. Cell growth by MTT test (Qiagen, Germany). The additional on-column digestion of gDNA was
MTT assay was performed to monitor the growth of HT29 cells applied with the aid of the RNase-free DNase Set (Qiagen, Germany).
exposed to the investigated compounds as described previously [15]. The quality and quantity of isolated RNA were estimated using Nano­
Briefly, cells were seeded in 96-well tissue culture plates at the density of drop 2000c built-in software (Thermo Scientific, USA). The isolated
5,000 cells per well and were allowed to settle for 24 h at 37 ◦ C. Then, RNA was stored at − 80 ◦ C until use. The amount of mRNA used for
the cells were treated either with purified water (as a control) or with reverse transcription (RT) was always equal to 500 ng. RT was con­
solutions of tested antioxidants for 6, 24, and 72 h within the final range ducted using the RT2 First Strand kit according to the manufacturer’s
of concentrations from 0.01 μM to 10 mM. All cells were allowed to grow protocol. The obtained cDNA was immediately diluted in qPCR master
for the total incubation time of 72 h, then cell growth was estimated by mix using the RT2 SYBR Green kit according to the instructions supplied
MTT assay. The measurement of absorption of formazan solutions was by the manufacturer (Qiagen, Germany). All instructions are available
performed at 540 nm using a TECAN Infinite M200 plate reader. The online at https://www.qiagen.com/us/resources/resourcedetail?
effect of tested AA derivatives was calculated and expressed as a per­ id=f4b13eaa-884f-4357-abe6-1a5f9469bc32&lang=en. The prepared
centage of cell growth determined for control cells treated with an master mix was subsequently aliquoted over 96-well RT2 Profiler PCR
appropriate solvent only (% control). human oxidative stress array consisting of 84 human genes related to
oxidative stress response and 3 controls assessing RT efficiency, 3 pos­
2.5.2. Cellular antioxidant activity by CAA assay itive PCR controls, a gDNA contamination control as well as reference
The procedure and analysis of CAA test results have been previously genes (Supplementary Table S1). The RT-PCR was performed according
described in detail [16]. In brief, HT29 cells were seeded in 96-well to the protocol of the manufacturer using LightCycler 96 (Roche,
plates (30,000 cells per well) and were allowed to settle for 24 h at Switzerland). Data analysis was performed using software available on
37 ◦ C. Then, the cells were treated with aqueous solutions of investi­ the manufacturer’s website (https://dataanalysis.qiagen.com/pcr/arra
gated ascorbates for 1 h in the concentration range from 1 to 1000 μM. yanalysis.php). Data were normalized based on the geometric aver­
All steps of the CAA assay (Cell Biolabs) were performed according to aging of 3 best-performing reference genes (B2M, HPRT1, and RPLP0).
the manufacturer’s protocol available online (https://www.cellbiolabs. The relative changes in gene expression were calculated with the aid of
com/sites/default/files/STA-349-cellular-antioxidant-activity-assay-kit the comparative threshold cycle method (ΔΔCt) [20].
.pdf). The measurements of fluorescence were carried out every 5 min
for 60 min at 480/530 nm (ex./em.) with TECAN Infinite M200 plate 2.7. Statistical analysis
reader. Calculations of CAA units were performed as described earlier
[20]. Statistical analysis was carried out using Prism 8.0 software package
(GraphPad Software, Inc., USA); the level of statistical significance was
2.5.3. Genotoxicity and protection against DNA fragmentation by comet set at p-value <0.05. All results were expressed as means ± SD of three
assay independent experiments.
HT29 cells were seeded in 24-well tissue culture plates at the density
of 100,000 cells per well and were allowed to settle for 24 h at 37 ◦ C. For
genotoxicity testing, cells were treated for 24 h with tested ascorbates
dissolved in sterile ultra-pure water in the concentration range from 1 to

242
P. Jakubek et al. Free Radical Biology and Medicine 209 (2023) 239–251

3. Results at each tested temperature were CaA and iNaA, respectively. In the ABTS
test, the antioxidant activity of AA and its isomer iAA was the same at
3.1. Standard reduction potential and antioxidant power higher temperatures, while at room temperature, iAA turned out to be a
significantly better radical scavenger. In the DPPH test, AA exhibited
PT is a technique that allows monitoring of the oxidation of a com­ stronger antioxidant activity than iAA, regardless of the temperature.
pound in the bulk solution and as such, measuring the thermodynamic NaA was a better radical scavenger than its isomer iNaA in both tests at
parameter E0 of a given redox couple. The parameter E0 describes the any tested temperature. All p-values were gathered in Supplementary
compound’s ability to accept electrons and is strongly related to the Table S3.
presence of redox-active moieties in its structure. The lower the E0, the
better electron donor a molecule is, which means that it exhibits
3.3. Modulation of lipid peroxidation
stronger antioxidant (i.e., reducing) properties. According to PT mea­
surements carried out for studied ascorbates, the antioxidant activity
The impact of AA and its derivatives on the kinetics of lipid peroxi­
among the studied group of compounds was shown to increase in the
dation was used as another parameter describing their antiradical ac­
following order: AA < NaA < iAA < iNaA < CaA (Table 1, Fig. 1b).
tivity. The method was based on monitoring the initiation of lipid
Among tested compounds, the strongest antioxidant turned out to be
oxidation by the formation of primary lipid oxidation products. Specif­
CaA with E0 value almost 10-fold lower compared to other AA de­
ically in our study, we monitored the formation of conjugated dienes in
rivatives. The sodium salts of both isomers of AA exhibited stronger
the structure of UFAs build-in the structure of liposome PLs induced by
reducing properties than their parental forms, whereas the L-stereoiso­
the presence of haem iron. As shown in Fig. 2a, all tested compounds at
mers turned out to be weaker electron donors than their corresponding
concentration range of 20–75 μM significantly increased the reaction
D-stereoisomers.
rate of liposome PL oxidation in the presence of haem iron compared to
The AOP calculated based on DPV determinations combines three
the control (p-values are gathered in Supplementary Table S4). For all
significant factors that describe the electrooxidation at the surface of
tested compounds, there was a trend of accelerating the PL oxidation
WE: a value of the transferred charge Qa (area under the voltammetric
along with the increasing concentration of tested antioxidants. Howev­
curve, the higher it is, the stronger the antioxidant properties are), the
er, when the maximum reaction rate was reached, a further increase in
rate of charge transfer expressed by the time of its transfer t (the shorter it
the concentration of AA or its derivatives led to a decrease in the reac­
is, the faster the electrooxidation process) as well as the charge transfer
tion rate. Based on the obtained results, we found very strong and sta­
energy expressed by the Ep,a (the higher it is, the stronger the antioxi­
tistically significant correlations between the concentration of the tested
dant). Again, CaA exhibited the strongest antioxidant properties, here
compounds and the acceleration of lipid oxidation (Supplementary
taking into account both thermodynamic and kinetic aspects, since its
Table S5). Among tested compounds, the strongest accelerator of lipid
electrooxidation process was characterized by the highest values of Ip,a
peroxidation in the presence of haem iron turned out to be iAA in the
and Ep,a (Table 1). The values of AOP were shown to be positively
concentration range between 40 and 50 μM, while at the highest con­
correlated with the reducing properties of studied compounds (Pearson
centrations – its parent compound, AA (Fig. 2a, Supplementary
r = − 0.9762, p-value 0.0008).
Table S6). CaA turned out to be a significantly stronger accelerator of
lipid peroxidation compared only to NaA and solely at the lowest tested
concentration (Fig. 2a, Supplementary Table S6).
3.2. Antioxidant activity by ABTS and DPPH tests

Spectrophotometric tests used in this study were modified in a way to 3.4. Modulation of protein carbonylation
reflect also the kinetics of redox reactions [18]. Therefore, the antioxi­
dant activity was expressed as the stoichiometric n10 value that de­ To determine the ability of AA and its derivatives to protect proteins
scribes how many molecules of a radical were scavenged by one against oxidative damage, the system developed to study the liposome
molecule of an antioxidant within 10 min reaction time. Stoichiometric PL peroxidation was used, with one modification, namely a model
value n10 is calculated as a tangent of a linear relationship between protein (BSA) was added to the reaction mixture. After the oxidation
concentrations of reduced radical and studied compounds at three forced by the reaction mixture components, proteins were extracted,
different temperatures: room temperature (25 ◦ C), the average physio­ and carbonyl derivative content was measured. The obtained results
logical temperature of the human body (37 ◦ C), and elevated tempera­ showed that all tested AA derivatives, except CaA, slightly inhibited
ture (41 ◦ C), which in the case of humans may be accompanied by protein carbonyl formation in the whole tested concentration range
excessive ROS formation and requires medical intervention. As shown in (Fig. 2b). The strongest protective effect among tested compounds
Fig. 1c,e, C-vitamers and other studied AA derivatives were significantly showed AA, followed by NaA and iAA, especially at the highest tested
better scavengers of ABTS than of DPPH radicals, regardless of the concentrations (Fig. 2b). L-stereoisomers tended to show a stronger
temperature (Fig. 1d,f, p-values are gathered in Supplementary protective effect than D-stereoisomers and sodium salts of both isomers
Table S2). In both tests, the strongest and the weakest radical scavengers of AA exhibited weaker protective properties than their parental forms,

Table 1
The characterization of the oxidation (in the bulk solution by potentiometric titration, PT) and electrooxidation (at the surface of WE by differential pulse voltammetry,
DPV) of ascorbic acid and its derivatives. The values of standard reduction potential (E0) were calculated based on PT measurements carried out at 37 ◦ C. The values of
anodic peak potential (Ep,a), anodic peak current (Ip,a), charge passed under the anodic waves (Qa), antioxidant energy (AOE), time of reaction (t) and antioxidant
power (AOP) were determined by DPV at 25 ◦ C; potential scan rate v = 0.1 V s− 1, ε = 0.103 V. Results are presented as means ± SD. Three independent experiments
were performed for each compound and technique.
Compound E0 [V] Ep,a + ϵ [V] Ip,a [μA] Qa [μC⋅cm− 2] AOE [μJ⋅cm− 2] t [s] AOP [μW⋅cm− 2]

Method PT DPV DPV DPV DPV DPV DPV

AA 0.287 ± 0.002 0.233 ± 0.010 15.7 ± 1.60 16.6 ± 0.6 215.7 ± 18.7 5.5 ± 0.5 39.3 ± 0.8
NaA 0.272 ± 0.002 0.163 ± 0.005 15.6 ± 0.20 18.3 ± 0.7 153.3 ± 0.7 5.0 ± 0.3 30.7 ± 1.9
iAA 0.258 ± 0.002 0.218 ± 0.007 14.15 ± 0.01 16.1 ± 0.1 186.5 ± 1.4 5.8 32.9 ± 0.7
iNaA 0.246 ± 0.002 0.256 ± 0.010 11.26 ± 1.03 14.8 ± 0.3 217.6 ± 41.9 5.8 ± 0.6 37.7 ± 5.9
CaA 0.021 ± 0.002 0.266 ± 0.012 25.1 ± 2.60 41.7 ± 2.2 659.3 ± 38.1 6.1 ± 0.3 107.6 ± 0.9

243
P. Jakubek et al. Free Radical Biology and Medicine 209 (2023) 239–251

Fig. 1. Antioxidant activity of vitamin C, C-vitamers, and other common ascorbic acid derivatives determined by electrochemical and spectrophotometric ap­
proaches. a, Chemical structures of studied compounds. b, The antioxidant activity of studied compounds expressed as standard reduction potential (E0) determined
by potentiometric titration (PT) at 37 ◦ C and antioxidant power (AOP) calculated based on the differential pulse voltammetry (DPV) results at 25 ◦ C with a potential
scan rate v = 0.1 V s− 1. Results are presented as means ± SD of three independent measurements. Different letters indicate significant differences in one-way ANOVA
with Tukey’s post hoc test for E0 or Kruskal-Wallis test for AOP with statistical significance set at p-value <0.05. c-f, The relationship between scavenged ABTS or
DPPH radicals and the concentration of ascorbic acid and its derivatives at 25, 37, and 41 ◦ C. Panels d and f show corresponding n10 value axes. The results are
expressed as means ± SD of three independent experiments carried out in triplicates. The abbreviations used refer to: AA, L-ascorbic acid; iAA, D-isoascorbic acid;
CaA, calcium L-ascorbate; NaA, sodium L-ascorbate; iNaA, sodium D-isoascorbate.

244
P. Jakubek et al. Free Radical Biology and Medicine 209 (2023) 239–251

Fig. 2. Redox-related activities of vitamin C, C-vitamers, and other common ascorbic acid derivatives. a, The impact of ascorbic acid and its derivatives on the
modulation of phospholipid oxidation in the presence of haem iron expressed as a maximum reaction rate (Vmax). The results are presented as means ± SD of three
independent experiments. Different letters indicate significant differences in one-way ANOVA with Tukey’s post hoc test with the level of significance set at p-value
<0.05. b, The ability of tested ascorbic acid derivatives to protect proteins (BSA) from oxidative damage. Results represent means ± SD of three independent
measurements. Statistically significant differences were determined by one-way ANOVA with Dunnett’s test vs. C+ and marked as (*) p-value <0.05, (***) p-value
<0.001, (****) p-value <0.0001; C+, carbonylated proteins not treated with antioxidants, C-, native BSA. c, The impact of ascorbic acid derivatives on cellular
antioxidant activity in the HT29 cell line. The results are expressed as means ± SD of three independent experiments carried out in triplicate. Statistical analysis was
carried out by one-way ANOVA with Tukey test with the level of significance set at p-value <0.05. Different letters represent statistically significant differences
between specific treatments. d,e, The genotoxicity of ascorbic acid derivatives towards HT29 cells, and their antigenotoxic impact in HT29 cells exposed to H2 O2 . The
cells were treated with studied ascorbate derivatives for 24 h (genotoxicity) or for 24 h prior to exposure to H2 O2 (antigenotoxic activity) and then submitted to the
comet assay procedure. The results are expressed as the mean percentage of DNA in comet tail ± SD from three independent experiments carried out in duplicate and
compared with C-, negative control (cells treated with solvent only) and C+, positive control (exposure to 150 μM H2 O2 ). Statistically significant differences were
determined by one-way ANOVA with Dunnett’s test and marked as (*) p-value <0.05. f, The impact of studied ascorbic acid derivatives on the growth of HT29 cells
determined by MTT assay after 6, 24, and 72 h treatment with tested compounds. Cell growth is expressed relative to control non-treated cells whose growth is
regarded as 100%. Results are means of three independent experiments carried out in quadruplicate. Statistically significant differences were determined by one-way
ANOVA with Dunnett’s test and marked as (*) p-value <0.05, (**) p-value <0.01, (***) p-value <0.001, (****) p-value <0.0001. The abbreviations used refer to: AA,
L-ascorbic acid; iAA, D-isoascorbic acid; CaA, calcium L-ascorbate; NaA, sodium L-ascorbate; iNaA, sodium D-isoascorbate.

245
P. Jakubek et al. Free Radical Biology and Medicine 209 (2023) 239–251

however, statistically significant differences occurred only in the case of growth.


the highest tested concentrations (p-values can be found in Supple­
mentary Table S7). In the case of CaA, the content of protein carbonyl 3.8. Nutrigenomic analysis
derivatives increased indicating pro-oxidative effects, which was not
observed for other tested compounds. The impact of C-vitamers on the expression of redox-related genes
was studied with the use of microarrays consisting of 84 genes involved
3.5. Antioxidant activity by CAA assay in oxidative stress response and antioxidant defense systems. The com­
pounds selected for genomic analysis were: AA selected as a parent
CAA assay has been recognized as a more biologically relevant test to compound and representative of the whole group as well as a human
measure antioxidant activity compared to chemical tests since it in­ exogenous antioxidant performing important endogenous functions, and
volves the aspects of cellular absorption and metabolism of redox-active CaA chosen as the strongest antioxidant according to E0, AOP, n10 values
compounds [19,20]. As shown in Fig. 2c, the CAA values of investigated and CAA values at 500 and 1000 μM. The concentrations selected for cell
AA derivatives were low to moderate at a wide range of concentrations treatment reflect the physiologically relevant situation: 50 μM repre­
(reaching up to 200 μM). Within this range, no significant differences sents the average plasma concentration of AA in healthy individuals,
between AA derivates could be observed. Only at higher concentrations who consume fruits and vegetables daily, while 200 μM is the highest
(≥500 μM), the impact of studied compounds on the reducing capacity plasma concentration of AA that could be reached after the prolonged
of cells was noticeably enhanced. At both the highest concentrations supplementation not exceeding maximum tolerated dose [3]. Besides,
used (500 and 1000 μM), CaA prominently increased the CAA values of the latter concentration could be easily achieved in the gastrointestinal
HT29 cells compared to other ascorbate derivatives. At the highest cells directly exposed to ingested food.
tested concentration (1000 μM), CaA caused almost complete inhibition As shown in Fig. 3., the treatment with the lower concentration (50
of probe oxidation (83% in the range of maximum 100%). The second μM) of AA significantly downregulated the expression of only two genes,
strongest ascorbate derivative was NaA. The CAA values for other CYBB and RNF7 by 3.7-fold and 1.8-fold, respectively. These genes are
compounds did not, however, significantly differ from those determined involved in the cellular response to oxidative stress as well as ROS-
for NaA. Regarding the CAA values of compounds applied to HT29 cells generation mechanisms of immune response. Six other genes involved
at the highest concentration (1000 μM), the order of antioxidant activity in protection against oxidative stress (GPX1, GSTZ1, PDLIM1, PRDX5,
rises as follows: iAA < AA < iNaA < NaA < CaA. PRDX6, PRNP) showed a trend toward being downregulated. A statis­
tically significant, nonetheless slight (1.3-fold) upregulation by 50 μM
3.6. Genotoxicity and protection against oxidative DNA damage by comet AA was observed only for OXSR1 encoding a protein called oxidative-
assay stress responsive kinase 1. A trend towards upregulation was observed
for four other genes (PRDX1, PRDX3, SOD1, TXN) encoding proteins that
To determine whether AA derivatives could exert genotoxic effects in indirectly or directly participate in H2 O2 metabolism. In turn, 50 μM
HT29 cells as well as to check if they prevent H2 O2 -induced DNA frag­ CaA did not significantly affect the expression of any of the investigated
mentation, cells were first incubated with tested compounds for 24 h. genes. Only expression of two genes, MGST3 and PRDX4, involved in
When the treatment was finished, the cells were subjected either to the xenobiotic metabolism and H2 O2 reduction, showed a slight trend to­
comet assay procedure (in the case of genotoxicity determination) or to wards upregulation (1.4-fold). Thus, despite the presence of a common
1 h treatment with 150 μM H2 O2 (to assess protection against oxidative AA structure in both investigated derivatives, their impact on the tran­
genotoxic insult), followed by the comet assay procedure. scription of redox-related genes in HT29 cells was very different at 50
The results obtained using comet assay showed that neither low μM treatment.
(achievable with normal diet) nor high concentrations (achievable only The treatment of HT29 cells with the higher concentration (200 μM)
by pharmacological interventions) of AA derivatives induced any DNA of AA had a greater impact on the downregulation of RNF7 gene (2.3-
fragmentation in HT29 cells (Fig. 2d). Further verification of the ability fold) compared to the exposure to a lower concentration described
of the studied compounds to protect cells against H2 O2 -induced oxida­ above. Other downregulated genes were PTGS2 (2-fold decrease), as
tive DNA damage was conducted using physiologically relevant con­ well as PDLIM1 (1.7-fold decrease) and PRDX6 (1.4-fold decrease)
centrations, chosen based on the following criteria: average plasma involved in protection against oxidative stress. The upregulation upon
concentration of healthy individual (50 μM), moderate (100 μM), and cell treatment with 200 μM AA was observed for GCLC and GCLM (1.5-
intense oral supplementation (200 μM) [3]. At neither of these con­ fold and 1.75-fold, respectively), which encode two subunits of
centrations, AA derivatives protected cells against H2 O2 -induced DNA glutamate-cysteine ligase, the rate-limiting enzyme in glutathione syn­
fragmentation (Fig. 2e). In both variants of comet assay, in the case of thesis as well as for PRDX4 (peroxiredoxin 4) and TXN (thioredoxin),
non-treated cells (negative control, C-), the damage did not exceed 3 % indirectly involved in recycling of GSH from GSSG (upregulated by 1.4-
of DNA in a comet tail. In a second variant of the test, pre-treatment of fold and 1.9-fold, respectively). The expression of genes encoding
cells with higher concentrations of C-vitamers increased DNA frag­ cytosolic SOD1 and mitochondrial PRDX3 was increased 2.1-fold each,
mentation induced upon genotoxic treatment with H2 O2 , which in some both of which support the removal of ROS. In the case of 200 μM CaA
cases reached statistical significance. treatment, CCL5, encoding a protein that mediates inflammatory pro­
cesses was the only significantly upregulated gene (2.2-fold). Other
3.7. Cell growth by MTT test genes that showed a trend towards upregulation (approximately 2-fold)
were GCLM, PRDX1, PRDX3, and SOD1, all involved in ROS
As shown in Fig. 2f, shorter treatments (6 and 24 h) of HT29 cells detoxification.
with AA, iAA, CaA, and NaA did not significantly affect their growth in
the range of concentrations reaching up to 1 mM. Only after 72 h the cell 4. Discussion
growth was slightly inhibited, however, by no more than 20%. In
contrast, a notable (however, statistically insignificant) cell growth The antioxidant properties of AA and its derivatives (CaA, NaA) or
stimulation was observed after shorter treatments with iNaA, which isomers (iAA, iNaA) have been assumed to be alike and current research
decreased to basal level after 72 h. All compounds applied to the cells at on these compounds is focused mostly on their applicability in food
the highest dose, that is 10 mM, occurred to drastically reduce cell processing [21–24]. However, research aimed to explore their biological
growth regardless of incubation time. The only exception was 10 mM activity, beyond the minimum required for allowance to use them as
AA, which after the shortest treatment only slightly inhibited cell food additives, as well as complex characterization of their redox-related

246
P. Jakubek et al. Free Radical Biology and Medicine 209 (2023) 239–251

Fig. 3. Nutrigenomic activity of vitamin C and one of the C-vitamers – calcium ascorbate. a, Heatmap presenting modulation of 84 oxidative stress response and
antioxidant defense genes in HT29 cells after 24 h treatment with ascorbic acid (AA) and calcium ascorbate (CaA) at 50 or 200 μM concentrations. b, Relevant
changes in the expression of genes observed following the indicated treatment. The results are calculated based on three independent experiments. Data and sta­
tistical analysis were performed at QIAGEN’S GeneGlobe Data Analysis Center following the manufacturer’s instructions. Statistically significant changes (p-values
<0.05) are highlighted in grey. Full names of investigated genes can be found in Supplementary Table S1.

properties, has been lacking. Therefore, in this study we examined the well as for different green tea catechins [18,27]. In turn, the CAA assay,
hypothesis of whether the (electro)chemical properties of different AA which is considered a more biologically relevant method compared to
derivatives determine their antioxidant effects in a biological setting ABTS or DPPH tests, revealed that as long as HT29 cells were exposed to
using human colon adenocarcinoma HT29 cells as in vitro cellular model AA and its derivatives at concentrations achievable in plasma i.e. not
of the gastrointestinal tract [25]. higher than 200 μM [3], the impact on the antioxidative cellular barrier
The antioxidant activity of studied AA derivatives was evaluated was mostly indistinguishable. However, vitamin C while orally ingested
using different methodological approaches. Two electrochemical pa­ can reach much higher concentrations in the intestinal lumen [30],
rameters describing compounds’ reducing properties were determined which further accumulate in high concentrations in tissues such as, e.g.,
by electrochemical methods: (i) E0, the most commonly used parameter liver, pancreas, or kidneys [31]. Therefore, such exposures were also
characterizing the thermodynamic behaviour of redox-active com­ investigated. In line with (electro)chemical parameters, CaA again
pounds [26], and (ii) AOP, combining both thermodynamics and ki­ appeared to exert the strongest antioxidant activity but only when
netics of redox processes [27,28]. Both parameters were previously applied to cells at a concentration of 500 μM or higher (Fig. 2c). In
successfully applied as predictors of bioactivity of redox-active com­ general, the majority of strong and statistically significant correlations
pounds [18,27,28]. According to E0 and AOP, the strongest antioxidant between CAA values and (electro)chemical parameters (E0, AOP, and n10
activity among tested compounds was exhibited by CaA (Fig. 1b and values) occurred for higher (200–500 μM) concentrations of AA and its
Table 1), which is not surprising as its chemical structure contains two derivatives (Fig. 4, p-values are gathered in Supplementary Table S8). It
ascorbate moieties that are released as a result of dissociation in an could be assumed that such high concentrations of physiologically
aqueous environment. In line with the electrochemical parameters, CaA relevant antioxidants may override the redox homeostatic capacity of
was also the strongest scavenger of both ABTS and DPPH radicals, cells, which we previously observed also in the case of catechins [18].
commonly used to assess the antioxidant activity of plant foods and After the assessment of antioxidant activity using three different
beverages [29]. Indeed, electrochemical parameters and n10 values were approaches, in the next step, we examined how AA and its derivatives
strongly correlated as shown in Fig. 4. Such strong relationships between affect the oxidation processes of biomolecules, as these are biologically
electrochemical parameters (E0, AOP, which express thermodynamic relevant targets of ROS. Initially, the kinetics of lipid peroxidation and
behaviour of a compound) and n10 values (involving the aspect of redox protein carbonylation was investigated in a cell-free system. For this
reaction kinetics) were also shown in our previous studies on purpose, we used haem iron as a physiological and effective initiator of
redox-related properties of different polyphenols, i.e. quercetin, nar­ lipid peroxidation [32]. Moreover, this form of iron, when consumed in
ingenin (aglycones), rutin, naringin (glycosides) or their mixtures, as excess, seems to be an important factor in the induction of intestinal

247
P. Jakubek et al. Free Radical Biology and Medicine 209 (2023) 239–251

iAA) showed the most potent protective effects when applied at a high
physiologically relevant concentration (100 μM) (Fig. 2b), with AA
turning out to be significantly stronger than both NaA and iAA (Sup­
plementary Table S7). At lower concentrations, again, mostly AA and
then NaA showed statistically significant albeit slight protection against
protein carbonylation. Interestingly, it has been shown that 5-week
supplementation with vitamin C (400 mg/day, a form of ascorbate
was not specified) can indeed reduce the level of protein carbonylation
but only in healthy volunteers with low baseline ascorbate level (base­
line ascorbate ~30 μM). Individuals with normal baseline vitamin C
status (~50 μM) did not benefit from supplementation [39]. In contrast,
CaA, which occurred to be the strongest antioxidant in (electro)chemical
and cellular tests (Fig. 1b,d,f and 2c), showed an unexpected proox­
idative effect at most tested concentrations, with 30 μM concentration
exhibiting the most potent effects (Fig. 2b). Altogether, these results
show that the impact of AA and its derivatives on protection against
protein carbonylation is concentration-dependent, as well as that AA
derivatives are less effective than the parent compound. In line with our
findings, some reports have previously shown that AA can either
decrease (at 1 mM concentration) or increase (at 10 mM concentration)
levels of protein carbonylation [40,41]. In the latter case, it has been
suggested that ascorbic acid could serve as a potential source of protein
Fig. 4. (Electro)chemical parameters describing antioxidant activity are carbonylation due to its autoxidation to dehydroascorbic acid, which
strongly correlated with cellular antioxidant activity of high concentrations of can further react with amino groups in proteins and generate products of
ascorbic acid and its derivatives. Correlations between values of standard
Maillard reaction and finally, advanced glycation end products [40,42].
reduction potential (E0) as well as other electrochemical parameters (Ep,a, Ip,a,
Other biomolecules liable to oxidative damage are nucleic acids.
Qa, and AOP) and values expressing antioxidant activity of ascorbic acid and its
Therefore, the ability of physiologically-relevant concentrations of AA
derivatives determined by chemical (n10 values for ABTS and DPPH tests) and
biological (CAA) approaches examined using Pearsons’s coefficients. The larger and its derivatives to provide protection against H2 O2 -induced oxidation
the dot, the stronger the correlation; the dark blue colour indicates a strong of DNA in HT29 cells was examined by comet assay. So far numerous
positive correlation, dark red colour shows a strong negative correlation. observational as well as interventional studies showed that vitamin C at
plasma concentrations exceeding 50 μM protects the integrity of DNA
oxidative stress and potentially contribute to the development of some under oxidative stress conditions in vivo [43]. However, somewhat
diseases including colorectal cancer [33]. The obtained data indicated surprisingly, in this study preincubation of HT29 cells with high albeit
that in the presence of haem iron, lower concentrations of AA and its physiologically-relevant concentrations of C-vitamers, i.e. AA, NaA, and
derivatives accelerated PL oxidation. Such results are not surprising CaA, followed by short genotoxic H2 O2 treatment resulted in increased
since ascorbate is known to act in a concentration-dependent manner as levels of DNA fragmentation (Fig. 2e). Such an effect can be explained by
not only an antioxidant but also a propagator of prooxidant effects on the already discussed prooxidative behaviour of ascorbates resulting
lipids in the presence of transition metal ions such as iron or copper as from their involvement in Fenton reactions (reactions R1-R3). The
shown by reactions 1–3: previously reported DNA damage induced in cellular systems has been
usually observed for millimolar concentrations of ascorbate [44,45].
Fe2+ + H2 O2 (or LOOH) → Fe3+ + HO• (or LO• ) + OH− (R1) The ability of ascorbate to prevent oxidative DNA damage in HT29 cells
has been also earlier reported [46]. In the latter case, experimental
Fe3+ + H2 O2 (or LOOH) → Fe2+ + HOO• (or LOO• ) + H+ (R2) conditions and concentrations used markedly differed from those
applied in our study, i.e., vitamin C-treated HT29 cells were kept in
Fe3+ + AA → Fe2+ + AA•− + 2H+ (R3) suspension during the experiment, and part of the cells was treated with
3+
Reaction (R1) leading to the formation of Fe is markedly faster only 10 μM H2 O2 for the last 10 min of the total 60 min vitamin C
than reaction (R2), in which Fe2+ becomes restored (reaction rate co­ treatment with this antioxidant [46]. In such circumstances, the direct
efficients k equal 5.7 × 102 M− 1s− 1 and 2.6 × 10− 3 M− 1s− 1, respec­ chemical neutralization of H2 O2 by AA could occur not related to the
tively) [34,35]. The presence of AA and its derivatives contributes to the redox status of the cells as investigated in our system. Although there is
acceleration of Fe2+ regeneration, as shown by the reaction (R3). Ulti­ much evidence that vitamin C may protect DNA against oxidative
mately, this propagates reaction (R1), leads to increased ROS produc­ damage, there is still a lot of inconclusive and contradictory data. The
tion, and results in the acceleration of lipid oxidation. Furthermore, our protective effect of vitamin C against DNA damage may depend on the
data showed that AA and iAA had a greater contribution to increased PL dose and incubation time and, as suggested by some researchers, it re­
oxidation when compared to their sodium salts (NaA and iNaA). Indeed, quires further insight [35].
such an effect could be attributed to their acidic forms since it has been In this study, the HT29 cell line was used as in vitro model of the
shown that a slight decrease in pH value could increase lipid oxidation intestinal epithelium, however, some data interpretations may also refer
mediated by hemoglobin in cod microsomal membranes [36]. This to the malignant origin of these cells [9,25]. It is widely recognized that
observation can be related to the pH-induced changes in hemoglobin quickly proliferating cancer cells are characterized by enhanced ROS
structure leading to the acceleration of lipid oxidation [37]. In turn, generation and upregulated endogenous antioxidant capacity, which
when the maximum lipid oxidation rate was achieved, a further increase creates a microenvironment favourable for the further proliferation of
in the concentration of AA derivatives led to a decrease in the oxidation cancer cells [47,48]. There is also evidence from in vivo studies that a
rate. This effect may be related to the fact that at high concentrations surplus of exogenously administered antioxidants can support mela­
reducing compounds are present in sufficient amounts to also scavenge noma and lung cancer progression [49,50]. In our study, iNaA turned
ROS generated in a reaction mixture [38]. out to be the only compound that showed a trend of notable enhance­
In the case of protein carbonylation, AA and its two derivatives (NaA, ment of HT29 cell growth, even though its redox-related activity did not
stand out among other AA derivatives. Seeing that no evidence of the

248
P. Jakubek et al. Free Radical Biology and Medicine 209 (2023) 239–251

tumorigenic impact of iNaA has been noted in the past in vivo studies between AA and GSH in plants and animals, including humans, in terms
[51,52], the observed trend should be replicated using other (colon) of the maintenance of redox homeostasis [59,60]. Increased expression
cancer cell lines before drawing any firm conclusions. In contrast, of GCL, one of the subunits of enzyme participating in the synthesis of
millimolar concentrations of AA derivatives caused significant inhibi­ GSH, may also suggest that AA induced enhanced preservation of the
tion of cell growth and such an effect has been thoroughly documented GSH pool. Upregulation of TXN may also indirectly affect the regener­
in scientific literature [45,53,54]. The ascorbate-induced inhibition of ation of GSH, since TXN is utilized by PRDXs, which are other enzymes
cell growth may be a result of disrupted redox homeostasis towards engaged in the neutralization of H2 O2 [61]. Here, mitochondrial PRDX3
oxidative stress since ascorbate at millimolar concentrations (i) un­ as well as cytosolic PRDX1 and PRDX4 were upregulated, while other
dergoes auto-oxidation and triggers the generation of H2 O2 that can be cytosolic isoforms – PRDX5 and PRDX6, were downregulated. Based on
further transformed into much more reactive hydroxyl radical (HO• ) the obtained results, it is hard to notice any clear pattern of changes in
and (ii) depletes intracellular GSH and NADPH pools [55]. Such high gene expression, which would suggest definite more antioxidant or
concentrations of ascorbates can be achieved solely through intravenous pro-oxidant effects.
injection, at least in the bloodstream [56]. Interestingly, AA and CaA did not occur to support cellular redox
Finally, we looked at the impact of AA on cellular redox-related homeostasis unidirectionally. In contrast to AA, the majority of changes
function at the genomic level for two compounds that behaved differ­ in the expression of redox-related genes observed after the treatment
ently in previous experiments – AA and CaA – despite obvious chemical with CaA, missed to reach statistical significance. Such an observation at
structural similarity (Fig. 3a). Since these experiments were intended to first may seem rather counterintuitive, seeing that CaA occurred to be a
spot even subtle changes in the patterns of gene expression, alterations significantly stronger antioxidant in terms of its (electro)chemical
in the expression of genes, which failed slightly to reach statistical sig­ properties, which stood out among the rest of the tested derivatives.
nificance (p-values between 0.05 and 0.1) were also included in Fig. 3b However, previously we made similar observations for glutathione,
to indicate a trend in gene expression patterns. The reason was that in whose activity was weaker compared to the group of apparently stron­
cells treated with physiologically relevant concentrations of non-toxic ger reducing agents – catechins [18]. In that case, the treatment of HT29
dietary antioxidants, minimal influence on transcriptome can be ex­ cells with glutathione affected different sets of redox-related genes than
pected. In contrast, such major specific alterations in gene expression catechins, whose low, physiologically-relevant concentrations had an
may be seen, for example, in drug research. As depicted in Figs. 3 and 5, impact on the expression of solely 3 genes, which was diminished after
the treatment of cells with a low concentration of AA (considered treatments with higher concentration [18]. It could be hypothesized that
optimal in the context of plasma concentrations) caused significant the lack of notable effects on the expression of the antioxidant defense
downregulation of CYBB, which makes one of the subunits of one of the system after treatment with strong reducing agents such as CaA could be
major ROS-generating enzymes – NADPH oxidase [57]. Such results derived from the proper redox balance maintained in cells after treat­
could suggest a decrease in the generation of O•−2 , and thus alleviation of ment, so the expression of antioxidant genes was no longer needed. The
oxidative stress. On the other hand, a trend towards decreased expres­ observed diversified cellular response to the treatment with AA and its
sion of SOD3, which acts in the extracellular milieu, might suggest either calcium salt rather points to the fine regulation of endogenous systems
lowered antioxidant protection or no need to even induce it. At the same of antioxidant protection preventing both oxidative and reductive stress
time, the expression of SOD1, which converts O•− 2 to less reactive H2 O2 , occurrence [62]. The explanation of a similar phenomenon has been
was significantly upregulated, but only after treatment with a higher previously proposed by our group in the case of sulfiredoxin-mediated
concentration of AA. The possible enhanced production of H2 O2 did not maintenance of redox homeostasis in response to the treatment with
seem to be counterbalanced by the changed expression of other genes. catechins [63].
This would suggest the increased preservation of H2 O2 , which besides
being one of ROS, is known to mediate intracellular signalling [58].
Furthermore, it is commonly known that there is a strong interplay

Fig. 5. Nutrigenomic effects of vitamin C. a, Cellular response to oxidative stress upon treatment with vitamin C represented by ascorbic acid (AA) and calcium
ascorbate (CaA) can be modulated at the level of gene expression. b, Genes affected by the treatment of HT29 cells with AA and CaA are involved in numerous
pathways related to ROS neutralization, antioxidant protection, and cellular metabolism. Genes whose expression was significantly changed in response to vitamin C
treatments are written in bold (p-value <0.05), other affected genes refer to p-values between 0.05 and 0.1. The abbreviations refer to: CAT, catalase; CCL5, che­
mokine (C–C motif) ligand 5; CYBA, cytochrome b-245, alpha polypeptide; CYBB, cytochrome b-245, beta polypeptide; Cys, L-cysteine; DHA, dehydroascorbic acid;
FTH1, ferritin, heavy polypeptide 1; GCL, glutamate cysteine ligase; GCLC, glutamate-cysteine ligase, catalytic subunit; GCLM, glutamate-cysteine ligase, modifier
subunit; Glu, glutamic acid; Gly, glycine; GPX, glutathione peroxidase; GSH, glutathione; GSS, glutathione synthetase; GSSG, glutathione disulphide; GSTZ1,
glutathione transferase zeta 1; MGST3, microsomal glutathione S-transferase 3; NADP(H), nicotinamide adenine dinucleotide phosphate; OXSR1, oxidative-stress
responsive 1; PDLIM1, PDZ and LIM domain 1; PRDX, peroxiredoxin; PRNP, prion protein; PTGS2, prostaglandin-endoperoxide synthase 2 (prostaglandin G/H
synthase and cyclooxygenase); RNF7, ring finger protein 7; SOD, superoxide dismutase; SVCT, sodium-dependent vitamin C transporter; TXN, thioredoxin; TXNR,
thioredoxin reductase; VIMP, selenoprotein S.

249
P. Jakubek et al. Free Radical Biology and Medicine 209 (2023) 239–251

5. Conclusions [2] Z.A.M. Daud, A. Ismail, B. Sarmadi, Ascorbic Acid: Physiology and Health Effects,
first ed., Elsevier Ltd., 2015 https://doi.org/10.1016/B978-0-12-384947-2.00045-
3.
In conclusion, our study shows that integrating both electrochemical [3] S.J. Padayatty, H. Sun, Y. Wang, H.D. Riordan, S.M. Hewitt, A. Katz, R.A. Wesley,
and biological approaches is essential to comprehensively understand M. Levine, Vitamin C pharmacokinetics: implications for oral and intravenous use,
and potentially predict the behavior of redox-active compounds within Ann. Intern. Med. 140 (2004) 533–538, https://doi.org/10.7326/0003-4819-140-
7-200404060-00010.
biological systems. This integration is crucial because electrochemistry [4] S.K. Chang, A. Ismail, Z.A.M. Daud, Ascorbic acid: properties, determination and
underpins the chemical foundation of redox-related biological activities uses, in: B. Caballero, P.M. Finglas, F. Toldra (Eds.), Encycl. Food Heal., first ed.,
of antioxidants. We found that the distinctive behaviour of the studied Academic Press, 2015, pp. 275–284, https://doi.org/10.1016/B978-0-12-384947-
2.00044-1.
ascorbate derivatives in cellular antioxidant activity occurred solely at [5] Scientific Opinion on the re-evaluation of ascorbic acid (E 300), sodium ascorbate
higher concentrations achievable either in the intestinal lumen (being in (E 301) and calcium ascorbate (E 302) as food additives, EFSA J. 13 (2015), 4087,
contract with ingested food) or in various tissues (e.g., liver) as a result https://doi.org/10.2903/j.efsa.2015.4087.
[6] Scientific Opinion on the re-evaluation of erythorbic acid (E 315) and sodium
of ascorbate accumulation. The CAA values, obtained for higher con­ erythorbate (E 316) as food additives, EFSA J. 14 (2016), 4360, https://doi.org/
centrations of ascorbates, were shown to be strongly correlated with 10.2903/j.efsa.2016.4360.
electrochemical and kinetic parameters describing redox processes. In [7] M. Carocho, P. Morales, I.C.F.R. Ferreira, Antioxidants: reviewing the chemistry,
food applications, legislation and role as preservatives, Trends Food Sci. Technol.
the case of modulation of oxidative damage, the differences between the 71 (2018) 107–120, https://doi.org/10.1016/j.tifs.2017.11.008.
studied ascorbate derivatives were observed already at low concentra­ [8] J. Du, J.J. Cullen, G.R. Buettner, Ascorbic acid: chemistry, biology and the
tions that could be considered physiologically relevant. At the nutrige­ treatment of cancer, Biochim. Biophys. Acta Rev. Canc 1826 (2012) 443–457,
https://doi.org/10.1016/j.bbcan.2012.06.003.
nomic level, the distinct cellular response observed in the treatment
[9] D. Martínez-Maqueda, B. Miralles, I. Recio, HT29 cell line, in: K. Verhoeckx,
with AA and its calcium salt, which happens to exhibit the highest P. Cotter, I. López-Expósito, C. Kleiveland, T. Lea, A. Mackie, T. Requena,
antioxidant potency among AA derivatives, may suggest fine regulation D. Swiatecka, H. Wichers (Eds.), The Impact of Food Bioactives on Health: In vitro
of redox homeostasis. This result warrants deeper investigation. Future and Ex vivoModels, 2015, https://doi.org/10.1007/978-3-319-16104-4_11.
[10] K. Lenaerts, F.K. Bouwman, W.H. Lamers, J. Renes, E.C. Mariman, Comparative
research should intend to confirm these findings in more in vitro and proteomic analysis of cell lines and scrapings of the human intestinal epithelium,
then in vivo models to verify whether different redox-related properties BMC Genom. 8 (2007) 1–14, https://doi.org/10.1186/1471-2164-8-91.
of AA derivatives can have, so far omitted, health implications. If [11] C. Gorrini, I.S. Harris, T.W. Mak, Modulation of oxidative stress as an anticancer
strategy, Nat. Rev. Drug Discov. 12 (2013) 931–947, https://doi.org/10.1038/
confirmed, these findings may be relevant to the designing of nutra­ nrd4002.
ceuticals, dietary supplements, or even trials investigating the effec­ [12] K. Parchem, B. Kusznierewicz, T. Chmiel, P. Maciołek, A. Bartoszek, Profiling and
tiveness of vitamin C in cancer therapies. qualitative assessment of enzymatically and thermally oxidized egg yolk
phospholipids using a two-step high-performance liquid chromatography protocol,
J. Am. Oil Chem. Soc. 96 (2019) 693–706, https://doi.org/10.1002/aocs.12218.
Author contributions [13] S. Azouzi, H. Santuz, S. Morandat, C. Pereira, F. Côté, O. Hermine, K. El Kirat,
Y. Colin, C. Le Van Kim, C. Etchebest, P. Amireault, Antioxidant and membrane
binding properties of serotonin protect lipids from oxidation, Biophys. J. 112
Conceptualization: A.B.; Formal analysis: P.J.; Investigation: P.J., K. (2017) 1863–1873, https://doi.org/10.1016/j.bpj.2017.03.037.
S., M.K., M.A., K.P., I.K.-M.; Methodology: M.K., B.K., W.C.; Visualiza­ [14] S. Kupper, I. Kłosowska-Chomiczewska, P. Szumała, Collagen and hyaluronic acid
tion: P.J.; Roles/Writing - original draft: P.J., K.P., I.K.-M., Writing - hydrogel in water-in-oil microemulsion delivery systems, Carbohydr. Polym. 175
(2017) 347–354, https://doi.org/10.1016/j.carbpol.2017.08.010.
review & editing: P.J., K.P., I.K.-M., A.B., Funding acquisition J.N., Re­ [15] I. Koss-Mikołajczyk, B. Kusznierewicz, J. Namieśnik, A. Bartoszek, Juices from non-
sources: A.B.; Supervision: A.B. typical edible fruits as health-promoting acidity regulators for food industry, LWT
64 (2015) 845–852, https://doi.org/10.1016/j.lwt.2015.06.072.
[16] I. Koss-Mikołajczyk, B. Kusznierewicz, W. Wiczkowski, T. Sawicki, A. Bartoszek,
The comparison of betalain composition and chosen biological activities for
Declaration of competing interests differently pigmented prickly pear (Opuntia ficus-indica) and beetroot (Beta
vulgaris) varieties, Int. J. Food Sci. Nutr. 70 (2019) 442–452, https://doi.org/
The authors declare that they have no known competing financial 10.1080/09637486.2018.1529148.
[17] I. Koss-Mikołajczyk, B. Kusznierewicz, A. Bartoszek, The relationship between
interests or personal relationships that could have appeared to influence phytochemical composition and biological activities of differently pigmented
the work reported in this paper. varieties of berry fruits; comparison between embedded in food matrix and isolated
anthocyanins, Foods 8 (2019) 646, https://doi.org/10.3390/foods8120646.
[18] M. Baranowska, K. Suliborska, W. Chrzanowski, B. Kusznierewicz, J. Namieśnik,
Acknowledgments A. Bartoszek, The relationship between standard reduction potentials of catechins
and biological activities involved in redox control, Redox Biol. 17 (2018) 355–366,
https://doi.org/10.1016/j.redox.2018.05.005.
This work was funded by the National Science Centre (Poland)
[19] I. Koss-Mikołajczyk, M. Baranowska, J. Namieśnik, A. Bartoszek, Determination of
through the MAESTRO 6 grant programme (application number 2014/ antioxidant activity of phytochemicals in cellular models by fluorescence/
14/A/ST4/00640). Part of the findings included in this manuscript luminescence methods, Postepy Hig. Med. Dosw. (Online) 71 (2017) 602–617,
(results of spectrophotometic tests, MTT, CAA, comet assays, and https://doi.org/10.5604/01.3001.0010.3841.
[20] K.L. Wolfe, R.H. Liu, Cellular Antioxidant Activity (CAA) assay for assessing
microarrays analysis) has been included in the doctoral thesis of P. antioxidants, foods, and dietary supplements, J. Agric. Food Chem. 55 (2007)
Jakubek entitled “Chemical and biological evaluation of antioxidant 8896–8907, https://doi.org/10.1021/jf0715166.
activity of endogenous redox-active compounds compared to plant- [21] O. Gimeno, I. Astiasara, J. Bello, Calcium ascorbate as a potential partial substitute
for NaCl in dry fermented sausages : efect on colour, texture and hygienic quality at
derived exogenous antioxidants” [2022, Gdańsk University of Technol­ different concentrations, Meat Sci. 57 (2001) 23–29, https://doi.org/10.1016/
ogy], the electronic version of which was published in the open, insti­ s0309-1740(00)00070-x.
tutional, digital repository of Gdańsk University of Technology, [22] S. Mola, A. Uthairatanakij, V. Srilaong, S. Aiamla-or, P. Jitareerat, Impacts of
sodium chlorite combined with calcium chloride, and calcium ascorbate on
Pomeranian Digital Library. microbial population, browning, and quality of fresh-cut rose apple, Agric. Nat.
Resour. 50 (2016) 331–337, https://doi.org/10.1016/j.anres.2016.12.001.
[23] D. Kowalczyk, W. Kazimierczak, E. Zięba, M. Mężyńska, M. Basiura-Cembala,
Appendix A. Supplementary data
S. Lisiecki, M. Karaś, B. Baraniak, Ascorbic acid- and sodium ascorbate-loaded
oxidized potato starch films: comparative evaluation of physicochemical and
Supplementary data to this article can be found online at https://doi. antioxidant properties, Carbohydr. Polym. 181 (2018) 317–326, https://doi.org/
org/10.1016/j.freeradbiomed.2023.10.400. 10.1016/j.carbpol.2017.10.063.
[24] B.D. Howes, L. Milazzo, E. Droghetti, M. Nocentini, G. Smulevich, Addition of
sodium ascorbate to extend the shelf-life of tuna meat fish: a risk or a benefit for
References consumers? J. Inorg. Biochem. 200 (2019), 110813 https://doi.org/10.1016/j.
jinorgbio.2019.110813.
[25] G. Krishna, G. Gopalakrishnan, Alternative in vitro models for safety and toxicity
[1] D.A. Bender, A Dictionary of Food and Nutrition, third ed., Oxford University Press,
evaluation of nutraceuticals, in: R. Gupta, R. Lall, A. Srivastava (Eds.),
2014 https://doi.org/10.1093/acref/9780199234875.001.0001.

250
P. Jakubek et al. Free Radical Biology and Medicine 209 (2023) 239–251

Nutraceuticals Effic. Saf. Toxic., 2016, pp. 355–385, https://doi.org/10.1016/ [45] A. Ghanem, A.M. Melzer, E. Zaal, L. Neises, D. Baltissen, O. Matar, H. Glennemeier-
B978-0-12-802147-7.00027-9. Elsevier Inc. Marke, F. Almouhanna, J. Theobald, M.A. Abu el Maaty, C. Berkers, S. Wölfl,
[26] J. Sochor, J. Dobes, O. Krystofova, B. Ruttkay-Nedecky, P. Babula, M. Pohanka, Ascorbate kills breast cancer cells by rewiring metabolism via redox imbalance and
T. Jurikova, O. Zitka, V. Adam, B. Klejdus, R. Kizek, Electrochemistry as a tool for energy crisis, Free Radic. Biol. Med. 163 (2021) 196–209, https://doi.org/
studying antioxidant properties, Int. J. Electrochem. Sci. 8 (2013) 8464–8489. 10.1016/j.freeradbiomed.2020.12.012.
[27] M. Baranowska, Z. Koziara, K. Suliborska, W. Chrzanowski, M. Wormstone, [46] R. Kontek, B. Kontek, K. Grzegorczyk, Vitamin C modulates DNA damage induced
J. Namieśnik, A. Bartoszek, Interactions between polyphenolic antioxidants by hydrogen peroxide in human colorectal adenocarcinoma cell lines (HT29)
quercetin and naringenin dictate the distinctive redox-related chemical and estimated by comet assay in vitro, Arch. Med. Sci. 9 (2013) 1006–1012, https://
biological behaviour of their mixtures, Sci. Rep. 11 (2021) 1–18, https://doi.org/ doi.org/10.5114/aoms.2013.39791.
10.1038/s41598-021-89314-0. [47] K.F. Zahra, R. Lefter, A. Ali, E. Abdellah, C. Trus, A. Ciobica, D. Timofte, The
[28] M. Baranowska, K. Suliborska, V. Todorovic, B. Kusznierewicz, W. Chrzanowski, involvement of the oxidative stress status in cancer pathology: a double view on
S. Sobajic, A. Bartoszek, Interactions between bioactive components determine the role of the antioxidants, Oxid. Med. Cell. Longev. 2021 (2021), 9965916,
antioxidant, cytotoxic and nutrigenomic activity of cocoa powder extract, Free https://doi.org/10.1155/2021/9965916.
Radic. Biol. Med. 154 (2020) 48–61, https://doi.org/10.1016/j. [48] J.D. Hayes, A.T. Dinkova-Kostova, K.D. Tew, Oxidative stress in cancer, Cancer Cell
freeradbiomed.2020.04.022. 38 (2020) 167–197, https://doi.org/10.1016/j.ccell.2020.06.001.
[29] A. Floegel, D.O. Kim, S.J. Chung, S.I. Koo, O.K. Chun, Comparison of ABTS/DPPH [49] V.I. Sayin, M.X. Ibrahim, E. Larsson, J.A. Nilsson, P. Lindahl, M.O. Bergo,
assays to measure antioxidant capacity in popular antioxidant-rich US foods, Antioxidants accelerate lung cancer progression in mice, Sci. Transl. Med. 6
J. Food Compos. Anal. 24 (2011) 1043–1048, https://doi.org/10.1016/j. (2014), https://doi.org/10.1126/scitranslmed.3007653, 221ra15.
jfca.2011.01.008. [50] K. Le Gal, M.X. Ibrahim, C. Wiel, V.I. Sayin, M.K. Akula, C. Karlsson, M.G. Dalin, L.
[30] J. Lykkesfeldt, P. Tveden-Nyborg, The pharmacokinetics of vitamin C, Nutrients 11 M. Akyürek, P. Lindahl, J. Nilsson, M.O. Bergo, Antioxidants can increase
(2019) 2412, https://doi.org/10.3390/nu11102412. melanoma metastasis in mice, Sci. Transl. Med. 7 (2015), 308re8, https://doi.org/
[31] S.J. Padayatty, M. Levine, Vitamin C: the known and the unknown and Goldilocks, 10.1126/scitranslmed.aad3740.
Oral Dis. 22 (2016) 463–493, https://doi.org/10.1111/odi.12446. [51] K. Inai, H. Akamizu, R. Eto, T. Nishida, K. Ohe, T. Kobuke, S. Nambu, K. Matsuki,
[32] C.U. Carlsen, J.K.S. Møller, L.H. Skibsted, Heme-iron in lipid oxidation, Coord. S. Tokuoka, Tumorigenicity study of sodium erythorbate administered orally to
Chem. Rev. 249 (2005) 485–498, https://doi.org/10.1016/j.ccr.2004.08.028. mice, Hiroshima J. Med. Sci. 38 (1989) 135–139.
[33] F. Guéraud, S. Taché, J.P. Steghens, L. Milkovic, S. Borovic-Sunjic, N. Zarkovic, [52] F.A. Andersen, Final report on the safety assessment of ascorbyl palmitate, ascorbyl
E. Gaultier, N. Naud, C. Héliès-Toussaint, F. Pierre, N. Priymenko, Dietary dipalmitate, ascorbyl stearate, erythorbic acid, and sodium erythorbate, Int. J.
polyunsaturated fatty acids and heme iron induce oxidative stress biomarkers and a Toxicol. 18 (1997) 1–26, https://doi.org/10.1177/109158189901800303.
cancer promoting environment in the colon of rats, Free Radic. Biol. Med. 83 [53] N. Shenoy, E. Creagan, T. Witzig, M. Levine, Ascorbic acid in cancer treatment: let
(2015) 192–200, https://doi.org/10.1016/j.freeradbiomed.2015.02.023. the Phoenix fly, Cancer Cell 34 (2018) 700–706, https://doi.org/10.1016/j.
[34] R. Romero, D. Contreras, M. Sepúlveda, N. Moreno, C. Segura, V. Melin, ccell.2018.07.014.
Assessment of a Fenton reaction driven by insoluble tannins from pine bark in [54] M. Uetaki, S. Tabata, F. Nakasuka, T. Soga, M. Tomita, Metabolomic alterations in
treating an emergent contaminant, J. Hazard Mater. 382 (2020), 120982, https:// human cancer cells by Vitamin C-induced oxidative stress, Sci. Rep. 5 (2015),
doi.org/10.1016/j.jhazmat.2019.120982. 13896, https://doi.org/10.1038/srep13896.
[35] J. Kaźmierczak-Barańska, K. Boguszewska, A. Adamus-Grabicka, B.T. Karwowski, [55] D. Mastrangelo, E. Pelosi, G. Castelli, F. Lo-Coco, U. Testa, Mechanisms of anti-
Two faces of vitamin C—antioxidative and pro-oxidative agent, Nutrients 12 cancer effects of ascorbate: cytotoxic activity and epigenetic modulation, Blood
(2020) 1501, https://doi.org/10.3390/nu12051501. Cells, Mol. Dis. 69 (2018) 57–64, https://doi.org/10.1016/j.bcmd.2017.09.005.
[36] M. Pazos, I. Medina, H.O. Hultin, Effect of pH on hemoglobin-catalyzed lipid [56] A.C. Carr, J. Cook, Intravenous vitamin C for cancer therapy - identifying the
oxidation in cod muscle membranes in vitro and in situ, J. Agric. Food Chem. 53 current gaps in our knowledge, Front. Physiol. 9 (2018), https://doi.org/10.3389/
(2005) 3605–3612, https://doi.org/10.1021/jf0403890. fphys.2018.01182.
[37] M.P. Richards, R. Aranda, C. He, G.N. Phillips, Effect of pH on structural changes in [57] H. Sumimoto, Structure, regulation and evolution of Nox-family NADPH oxidases
perch hemoglobin that can alter redox stability and heme affinity, J. Aquat. Food that produce reactive oxygen species, FEBS J. 275 (2008) 3249–3277, https://doi.
Prod. Technol. 18 (2009) 416–423, https://doi.org/10.1080/ org/10.1111/j.1742-4658.2008.06488.x.
10498850903223598. [58] H. Sies, Hydrogen peroxide as a central redox signaling molecule in physiological
[38] T. Van Hecke, J. Van Camp, S. De Smet, Oxidation during digestion of meat: oxidative stress: oxidative eustress, Redox Biol. 11 (2017) 613–619, https://doi.
interactions with the diet and Helicobacter pylori gastritis, and implications on org/10.1016/j.redox.2016.12.035.
human health, Compr. Rev. Food Sci. Food Saf. 16 (2017) 214–233, https://doi. [59] V. Montecinos, P. Guzmán, V. Barra, M. Villagrán, C. Muñoz-Montesino,
org/10.1111/1541-4337.12248. K. Sotomayor, E. Escobar, A. Godoy, L. Mardones, P. Sotomayor, C. Guzmán,
[39] J.L. Carty, R. Bevan, H. Waller, N. Mistry, M. Cooke, J. Lunec, H.R. Griffiths, The O. Vásquez, V. Gallardo, B. Van Zundert, M.R. Bono, S.A. Oñate, M. Bustamante, J.
effects of vitamin C supplementation on protein oxidation in healthy volunteers, G. Cárcamo, C.I. Rivas, J.C. Vera, Vitamin C is an essential antioxidant that
Biochem. Biophys. Res. Commun. 273 (2000) 729–735, https://doi.org/10.1006/ enhances survival of oxidatively stressed human vascular endothelial cells in the
bbrc.2000.3014. presence of a vast molar excess of glutathione, J. Biol. Chem. 282 (2007)
[40] T. Miyata, R. Inagi, K. Asahi, Y. Yamada, K. Horie, H. Sakai, K. Uchida, 15506–15515, https://doi.org/10.1074/jbc.M608361200.
K. Kurokawa, Generation of protein carbonyls by glycoxidation and lipoxidation [60] C.H. Foyer, G. Noctor, Ascorbate and glutathione: the heart of the redox hub, Plant
reactions with autoxidation products of ascorbic acid and polyunsaturated fatty Physiol. 155 (2011) 2–18, https://doi.org/10.1104/pp.110.167569.
acids, FEBS Lett. 437 (1998) 24–28, https://doi.org/10.1016/S0014-5793(98) [61] S.G. Rhee, H.A. Woo, Multiple functions of peroxiredoxins: peroxidases, sensors
01079-5. and regulators of the intracellular messenger H2O2, and protein chaperones,
[41] J. Chavez, W.G. Chung, C.L. Miranda, M. Singhal, J.F. Stevens, C.S. Maier, Site- Antioxidants Redox Signal. 15 (2011) 781–794, https://doi.org/10.1089/
specific protein adducts of 4-hydroxy-2(e)-nonenal in human THP-1 monocytic ars.2010.3393.
cells: protein carbonylation is diminished by ascorbic acid, Chem. Res. Toxicol. 23 [62] M. Kuczyńska, P. Jakubek, A. Bartoszek, More than just antioxidants: redox-active
(2010) 37–47, https://doi.org/10.1021/tx9002462. components and mechanisms shaping redox signalling network, Antioxidants 11
[42] L. Liu, L. Liu, J. Xie, M. Shen, Formation mechanism of AGEs in Maillard reaction (2022) 2403, https://doi.org/10.3390/antiox11122403.
model systems containing ascorbic acid, Food Chem. 378 (2022), 132108, https:// [63] P. Jakubek, J. Rajić, M. Kuczyńska, K. Suliborska, M. Heldt, K. Dziedziul,
doi.org/10.1016/j.foodchem.2022.132108. M. Vidaković, J. Namieśnik, A. Bartoszek, Beyond antioxidant activity: redox
[43] R.J. Sram, B. Binkova, P. Rossner, Vitamin C for DNA damage prevention, Mutat. properties of catechins may affect changes in the DNA methylation profile—the
Res., Fundam. Mol. Mech. Mutagen. 733 (2012) 39–49, https://doi.org/10.1016/j. example of SRXN1 gene, Antioxidants 12 (2023) 754, https://doi.org/10.3390/
mrfmmm.2011.12.001. antiox12030754.
[44] J. Rivière, J.L. Ravanat, J.R. Wagner, Ascorbate and H2O2 induced oxidative DNA
damage in Jurkat cells, Free Radic. Biol. Med. 40 (2006) 2071–2079, https://doi.
org/10.1016/j.freeradbiomed.2006.02.003.

251

You might also like